US11344546B2 - Benzamide compounds - Google Patents

Benzamide compounds Download PDF

Info

Publication number
US11344546B2
US11344546B2 US16/957,862 US201916957862A US11344546B2 US 11344546 B2 US11344546 B2 US 11344546B2 US 201916957862 A US201916957862 A US 201916957862A US 11344546 B2 US11344546 B2 US 11344546B2
Authority
US
United States
Prior art keywords
unsubstituted
compound
pharmaceutically acceptable
acceptable salt
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/957,862
Other languages
English (en)
Other versions
US20210009543A1 (en
Inventor
Joseph Robert Pinchman
Peter Qinhua HUANG
Kevin Duane Bunker
Rakesh Kumar Sit
Ahmed Abdi Samatar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Recurium Ip Holding LLC
Recurium IP Holdings LLC
Zeno Management Inc
Original Assignee
Recurium Ip Holding LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Recurium Ip Holding LLC filed Critical Recurium Ip Holding LLC
Priority to US16/957,862 priority Critical patent/US11344546B2/en
Assigned to RECURIUM IP HOLDINGS, LLC reassignment RECURIUM IP HOLDINGS, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: Zeno Royalties & Milestones, LLC
Assigned to Zeno Royalties & Milestones, LLC reassignment Zeno Royalties & Milestones, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZENO MANAGEMENT, INC.
Assigned to ZENO MANAGEMENT, INC. reassignment ZENO MANAGEMENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BUNKER, KEVIN DUANE, HUANG, Peter Qinhua, PINCHMAN, JOSEPH ROBERT, Samatar, Ahmed Abdi, SIT, RAKESH KUMAR
Publication of US20210009543A1 publication Critical patent/US20210009543A1/en
Application granted granted Critical
Publication of US11344546B2 publication Critical patent/US11344546B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5355Non-condensed oxazines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D205/00Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom
    • C07D205/02Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D205/04Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/12Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/46Oxygen atoms attached in position 4 having a hydrogen atom as the second substituent in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/56Nitrogen atoms
    • C07D211/58Nitrogen atoms attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/04Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/10Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms
    • C07D295/112Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/125Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/13Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/155Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present application relates to compounds that are Bcl-2 inhibitors and methods of using them to treat conditions characterized by excessive cellular proliferation, such as cancer and tumors, and viral infections such as infection with the human immunodeficiency virus (HIV).
  • HIV human immunodeficiency virus
  • Bcl-2 plays a role in cell death regulation, including apoptosis, necrosis and autophagy. Accordingly, alterations in Bcl-2 expression and function contribute to pathogenesis and progression of human cancers and tumors, and may facilitate certain viral infections such as HIV.
  • Some embodiments provide a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • compositions that can include an effective amount of one or more of compounds of Formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof.
  • Some embodiments described herein relate to a method for treating a cancer or a tumor described herein that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) to a subject having a cancer described herein.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) to a subject having a cancer described herein.
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for treating a cancer or a tumor described herein.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for treating a cancer or a tumor described herein.
  • Some embodiments described herein relate to a method for inhibiting replication of a malignant growth or a tumor described herein that can include contacting the growth or the tumor with an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof).
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • Other embodiments described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for inhibiting replication of a malignant growth or a tumor described herein.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for inhibiting replication of a malignant growth or a tumor described herein.
  • Some embodiments described herein relate to a method for treating a cancer described herein that can include contacting a malignant growth or a tumor described herein with an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof).
  • Other embodiments described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for treating a cancer described herein, wherein the use comprises contacting a malignant growth or a tumor described herein with the medicament.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for contacting a malignant growth or a tumor described herein, wherein the malignant growth or tumor is due to a cancer described herein.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • Some embodiments described herein relate to a method for inhibiting the activity of Bcl-2 that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) to a subject and can also include contacting a cell that expresses Bcl-2 with an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof).
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for inhibiting the activity of Bcl-2 in a subject or, in the manufacture of a medicament for inhibiting the activity of Bcl-2, wherein the use comprises contacting a cell that expresses Bcl-2.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for inhibiting the activity of Bcl-2 in a subject; or for inhibiting the activity of Bcl-2 by contacting a cell that expresses Bcl-2.
  • Some embodiments described herein relate to a method of ameliorating or treating a HIV infection that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof), and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof)
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for ameliorating or treating a HIV infection in a subject suffering from the HIV infection; or, in the manufacture of a medicament for ameliorating or treating a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for ameliorating or treating a HIV infection in a subject suffering from the HIV infection; or for ameliorating or treating a HIV infection by contacting a cell infected with HIV.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • an effective amount of a HIV latency reversing agent or a pharmaceutically acceptable salt thereof
  • Some embodiments described herein relate to a method of reducing the population of HIV infected cells that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof), and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or, for reducing the population of HIV infected cells, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or for reducing the population of HIV infected cells by contacting a cell infected with HIV.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or for reducing the population of HIV infected cells by contacting a cell infected with HIV.
  • Some embodiments described herein relate to a method of reducing the reoccurrence of a HIV infection in a subject that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or, for reducing the reoccurrence of a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • an effective amount of a HIV latency reversing agent or a pharmaceutically acceptable salt thereof
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or for reducing the reoccurrence of a HIV infection by contacting a cell infected with HIV.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or for reducing the reoccurrence of a HIV infection by contacting a cell infected with HIV.
  • Some embodiments described herein relate to a method of ameliorating or treating a HIV infection that can include administering an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • inventions described herein relate to the use of an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for ameliorating or treating a HIV infection; or, in the manufacture of a medicament for ameliorating or treating a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • Still other embodiments described herein relate to an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for ameliorating or treating a HIV infection in a subject suffering from the HIV infection; or for ameliorating or treating a HIV infection by contacting a cell infected with HIV.
  • Some embodiments described herein relate to a method of reducing the population of HIV infected cells that can include administering an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • inventions described herein relate to the use of an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the population of HIV infected cells; or, in the manufacture of a medicament for reducing the population of HIV infected cells, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • Still other embodiments described herein relate to an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or for reducing the population of HIV infected cells by contacting a cell infected with HIV.
  • Some embodiments described herein relate to a method of reducing the reoccurrence of a HIV infection in a subject that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof)
  • inventions described herein relate to the use of an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or, for reducing the reoccurrence of a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • Still other embodiments described herein relate to an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or for reducing the reoccurrence of a HIV infection by contacting a cell infected with HIV.
  • FIG. 1 shows example HIV latency reversing agents.
  • FIG. 2 shows example Bcl protein inhibitors.
  • FIG. 3 shows examples of compounds of the Formula (I).
  • FIGS. 4A, 4B and 4C show examples of compounds of the Formula (I).
  • FIG. 5 shows a plot illustrating HIV assay data for examples of compounds of the Formula (I).
  • Bcl-2 is a critical regulator of programmed cell death (apoptosis).
  • Bcl-2 belongs to the B cell lymphoma 2 (BCL-2) a family of proteins, which includes both pro-apoptotic proteins (such as Bak, Bax, Bim, Bid, tBid, Bad, Bik, PUMA, Bnip-1, Hrk, Bmf and Noxa) and anti-apoptotic proteins (such as Bcl-2, Bcl-X L , Bcl-W, Mcl-1 and Bcl-2A1).
  • pro-apoptotic proteins such as Bak, Bax, Bim, Bid, tBid, Bad, Bik, PUMA, Bnip-1, Hrk, Bmf and Noxa
  • anti-apoptotic proteins such as Bcl-2, Bcl-X L , Bcl-W, Mcl-1 and Bcl-2A1.
  • Bcl-2 inhibits apoptosis in
  • Bcl-2 Activation of the intrinsic apoptosis pathway (e.g., by cellular stress) inhibits Bcl-2, thus activating Bak and Bax.
  • These proteins facilitate mitochondrial outer membrane permeabilization, releasing cytochrome c and Smac. This initiates the caspase signaling pathway, ultimately resulting in cell death.
  • Dysregulation of Bcl-2 leads to sequestration of cell-death-promoting proteins, leading to evasion of apoptosis. This process contributes to malignancy, and facilitates cell survival under other disadvantageous conditions, such as during viral infection.
  • transcriptionally active HIV can incidentally generate a caspase protein fragment. This fragment binds to and activates the pro-apoptotic Bak.
  • this fragment also binds to, and is sequestered by, Bcl-2 reducing its effectiveness at inducing cell death. Inhibition of Bcl-2 disrupts sequestration of pro-apoptotic proteins, restoring apoptotic signaling, and promoting damaged cells to undergo programmed cell death. Therefore, Bcl-2 inhibition has the potential to ameliorate or treat cancers and tumors, as well as ameliorate or treat certain viral infections in combination with other agents.
  • the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), cycloalkyl(alkyl), heteroaryl(alkyl), heterocyclyl(alkyl), hydroxy, alkoxy, acyl, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, nitro, sulfenyl, sulfinyl, sulfonyl,
  • C a to C b in which “a” and “b” are integers refer to the number of carbon atoms in a group.
  • the indicated group can contain from “a” to “b”, inclusive, carbon atoms.
  • a “C 1 to C 4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH 3 —, CH 3 CH 2 —, CH 3 CH 2 CH 2 —, (CH 3 ) 2 CH—, CH 3 CH 2 CH 2 CH 2 —, CH 3 CH 2 CH(CH 3 )— and (CH 3 ) 3 C—. If no “a” and “b” are designated, the broadest range described in these definitions is to be assumed.
  • R groups are described as being “taken together” the R groups and the atoms they are attached to can form a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heterocycle.
  • R a and R b of an NR a R b group are indicated to be “taken together,” it means that they are covalently bonded to one another to form a ring:
  • alkyl refers to a fully saturated aliphatic hydrocarbon group.
  • the alkyl moiety may be branched or straight chain.
  • branched alkyl groups include, but are not limited to, iso-propyl, sec-butyl, t-butyl and the like.
  • straight chain alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl and the like.
  • the alkyl group may have 1 to 30 carbon atoms (whenever it appears herein, a numerical range such as “1 to 30” refers to each integer in the given range; e.g., “1 to 30 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 30 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated).
  • the alkyl group may also be a medium size alkyl having 1 to 12 carbon atoms.
  • the alkyl group could also be a lower alkyl having 1 to 6 carbon atoms.
  • An alkyl group may be substituted or unsubstituted.
  • alkylene refers to a bivalent fully saturated straight chain aliphatic hydrocarbon group.
  • alkylene groups include, but are not limited to, methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene and octylene.
  • An alkylene group may be represented by , followed by the number of carbon atoms, followed by a “*”. For example,
  • the alkylene group may have 1 to 30 carbon atoms (whenever it appears herein, a numerical range such as “1 to 30” refers to each integer in the given range; e.g., “1 to 30 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 30 carbon atoms, although the present definition also covers the occurrence of the term “alkylene” where no numerical range is designated).
  • the alkylene group may also be a medium size alkyl having 1 to 12 carbon atoms.
  • the alkylene group could also be a lower alkyl having 1 to 4 carbon atoms.
  • An alkylene group may be substituted or unsubstituted. For example, a lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group and/or by substituting both hydrogens on the same carbon with a C 3-6 monocyclic cycloalkyl group
  • alkenyl used herein refers to a monovalent straight or branched chain radical of from two to twenty carbon atoms containing a carbon double bond(s) including, but not limited to, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl and the like.
  • An alkenyl group may be unsubstituted or substituted.
  • alkynyl used herein refers to a monovalent straight or branched chain radical of from two to twenty carbon atoms containing a carbon triple bond(s) including, but not limited to, 1-propynyl, 1-butynyl, 2-butynyl and the like.
  • An alkynyl group may be unsubstituted or substituted.
  • cycloalkyl refers to a completely saturated (no double or triple bonds) mono- or multi-cyclic (such as bicyclic) hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro fashion. As used herein, the term “fused” refers to two rings which have two atoms and one bond in common. As used herein, the term “bridged cycloalkyl” refers to compounds wherein the cycloalkyl contains a linkage of one or more atoms connecting non-adjacent atoms.
  • Cycloalkyl groups can contain 3 to 30 atoms in the ring(s), 3 to 20 atoms in the ring(s), 3 to 10 atoms in the ring(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s).
  • a cycloalkyl group may be unsubstituted or substituted.
  • Examples of mono-cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • Examples of fused cycloalkyl groups are decahydronaphthalenyl, dodecahydro-1H-phenalenyl and tetradecahydroanthracenyl;
  • examples of bridged cycloalkyl groups are bicyclo[1.1.1]pentyl, adamantanyl and norbornanyl; and examples of spiro cycloalkyl groups include spiro[3.3]heptane and spiro[4.5]decane.
  • cycloalkenyl refers to a mono- or multi-cyclic (such as bicyclic) hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein).
  • Cycloalkenyl groups can contain 3 to 10 atoms in the ring(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s). When composed of two or more rings, the rings may be connected together in a fused, bridged or spiro fashion.
  • a cycloalkenyl group may be unsubstituted or substituted.
  • aryl refers to a carbocyclic (all carbon) monocyclic or multicyclic (such as bicyclic) aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings.
  • the number of carbon atoms in an aryl group can vary.
  • the aryl group can be a C 6 -C 14 aryl group, a C 6 -C 10 aryl group or a C 6 aryl group.
  • Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene.
  • An aryl group may be substituted or unsubstituted.
  • heteroaryl refers to a monocyclic or multicyclic (such as bicyclic) aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one or more heteroatoms (for example, 1, 2 or 3 heteroatoms), that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur.
  • heteroatoms for example, 1, 2 or 3 heteroatoms
  • the number of atoms in the ring(s) of a heteroaryl group can vary.
  • the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s), such as nine carbon atoms and one heteroatom; eight carbon atoms and two heteroatoms; seven carbon atoms and three heteroatoms; eight carbon atoms and one heteroatom; seven carbon atoms and two heteroatoms; six carbon atoms and three heteroatoms; five carbon atoms and four heteroatoms; five carbon atoms and one heteroatom; four carbon atoms and two heteroatoms; three carbon atoms and three heteroatoms; four carbon atoms and one heteroatom; three carbon atoms and two heteroatoms; or two carbon atoms and three heteroatoms.
  • heteroaryl includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring or at least two heteroaryl rings, share at least one chemical bond.
  • heteroaryl rings include, but are not limited to, furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine,
  • heterocyclyl or “heteroalicyclyl” refers to three-, four-, five-, six-, seven-, eight-, nine-, ten-, up to 18-membered monocyclic, bicyclic and tricyclic ring system wherein carbon atoms together with from 1 to 5 heteroatoms constitute said ring system.
  • a heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings.
  • the heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur and nitrogen.
  • a heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates.
  • oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates.
  • the rings When composed of two or more rings, the rings may be joined together in a fused, bridged or spiro fashion.
  • the term “fused” refers to two rings which have two atoms and one bond in common.
  • bridged heterocyclyl or “bridged heteroalicyclyl” refers to compounds wherein the heterocyclyl or heteroalicyclyl contains a linkage of one or more atoms connecting non-adjacent atoms.
  • spiro refers to two rings which have one atom in common and the two rings are not linked by a bridge.
  • Heterocyclyl and heteroalicyclyl groups can contain 3 to 30 atoms in the ring(s), 3 to 20 atoms in the ring(s), 3 to 10 atoms in the ring(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s).
  • any nitrogens in a heteroalicyclic may be quaternized.
  • Heterocyclyl or heteroalicyclic groups may be unsubstituted or substituted.
  • heterocyclyl or “heteroalicyclyl” groups include but are not limited to, 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1,2-dioxolane, 1,3-dioxolane, 1,4-dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3-dithiolane, 1,4-oxathiane, tetrahydro-1,4-thiazine, 2H-1,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, trioxane, hexahydro-1,3,5-triazine, imidazoline, imidazolidine, isoxazoline, isoxazol
  • spiro heterocyclyl groups examples include 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxaspiro[3.4]octane and 2-azaspiro[3.4]octane.
  • aralkyl and “aryl(alkyl)” refer to an aryl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2-phenylalkyl, 3-phenylalkyl and naphthylalkyl.
  • heteroarylkyl and “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylalkyl, 3-thienylalkyl, furylalkyl, thienylalkyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl and imidazolylalkyl and their benzo-fused analogs.
  • heteroalicyclyl(alkyl) and “heterocyclyl(alkyl)” refer to a heterocyclic or a heteroalicyclic group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and heterocyclyl of a (heteroalicyclyl)alkyl may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl(methyl), piperidin-4-yl(ethyl), piperidin-4-yl(propyl), tetrahydro-2H-thiopyran-4-yl(methyl) and 1,3-thiazinan-4-yl(methyl).
  • hydroxy refers to a —OH group.
  • alkoxy refers to the Formula —OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein.
  • R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein.
  • a non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy,
  • acyl refers to a hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) and heterocyclyl(alkyl) connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl and acryl. An acyl may be substituted or unsubstituted.
  • a “cyano” group refers to a “—CN” group.
  • halogen atom or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.
  • a “thiocarbonyl” group refers to a “—C( ⁇ S)R” group in which R can be the same as defined with respect to O-carboxy.
  • a thiocarbonyl may be substituted or unsubstituted.
  • An “O-carbamyl” group refers to a “—OC( ⁇ O)N(R A R B )” group in which R A and R B can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An O-carbamyl may be substituted or unsubstituted.
  • N-carbamyl refers to an “ROC( ⁇ O)N(R A )—” group in which R and R A can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An N-carbamyl may be substituted or unsubstituted.
  • An “O-thiocarbamyl” group refers to a “—OC( ⁇ S)—N(R A R B )” group in which R A and R B can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An O-thiocarbamyl may be substituted or unsubstituted.
  • N-thiocarbamyl refers to an “ROC( ⁇ S)N(R A )—” group in which R and R A can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An N-thiocarbamyl may be substituted or unsubstituted.
  • a “C-amido” group refers to a “—C( ⁇ O)N(R A R B )” group in which R A and R B can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • a C-amido may be substituted or unsubstituted.
  • N-amido refers to a “RC( ⁇ O)N(R A )—” group in which R and R A can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • R and R A can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An N-amido may be substituted or unsubstituted.
  • S-sulfonamido refers to a “—SO 2 N(R A R B )” group in which R A and R B can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An S-sulfonamido may be substituted or unsubstituted.
  • N-sulfonamido refers to a “RSO 2 N(R A )—” group in which R and R A can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • R and R A can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An N-sulfonamido may be substituted or unsubstituted.
  • An “O-carboxy” group refers to a “RC( ⁇ O)O—” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein.
  • An O-carboxy may be substituted or unsubstituted.
  • esters and C-carboxy refer to a “—C( ⁇ O)OR” group in which R can be the same as defined with respect to O-carboxy.
  • An ester and C-carboxy may be substituted or unsubstituted.
  • a “nitro” group refers to an “—NO 2 ” group.
  • a “sulfenyl” group refers to an “—SR” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • a sulfenyl may be substituted or unsubstituted.
  • a “sulfinyl” group refers to an “—S( ⁇ O)—R” group in which R can be the same as defined with respect to sulfenyl.
  • a sulfinyl may be substituted or unsubstituted.
  • a “sulfonyl” group refers to an “SO 2 R” group in which R can be the same as defined with respect to sulfenyl.
  • a sulfonyl may be substituted or unsubstituted.
  • haloalkyl refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl, tri-haloalkyl and polyhaloalkyl).
  • a halogen e.g., mono-haloalkyl, di-haloalkyl, tri-haloalkyl and polyhaloalkyl.
  • groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1-chloro-2-fluoromethyl, 2-fluoroisobutyl and pentafluoroethyl.
  • a haloalkyl may be substituted or unsubstituted.
  • haloalkoxy refers to an alkoxy group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di-haloalkoxy and tri-haloalkoxy).
  • a halogen e.g., mono-haloalkoxy, di-haloalkoxy and tri-haloalkoxy.
  • groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, 1-chloro-2-fluoromethoxy and 2-fluoroisobutoxy.
  • a haloalkoxy may be substituted or unsubstituted.
  • amino and “unsubstituted amino” as used herein refer to a —NH 2 group.
  • a “mono-substituted amine” group refers to a “—NHR A ” group in which R A can be an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein.
  • the R A may be substituted or unsubstituted.
  • a mono-substituted amine group can include, for example, a mono-alkylamine group, a mono-C 1 -C 6 alkylamine group, a mono-arylamine group, a mono-C 6 -C 10 arylamine group and the like.
  • Examples of mono-substituted amine groups include, but are not limited to, —NH(methyl), —NH(phenyl) and the like.
  • a “di-substituted amine” group refers to a “—NR A R B ” group in which R A and R B can be independently an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein.
  • R A and R B can independently be substituted or unsubstituted.
  • a di-substituted amine group can include, for example, a di-alkylamine group, a di-C 1 -C 6 alkylamine group, a di-arylamine group, a di-C 6 -C 10 arylamine group and the like.
  • Examples of di-substituted amine groups include, but are not limited to, —N(methyl) 2 , —N(phenyl)(methyl), —N(ethyl)(methyl) and the like.
  • “mono-substituted amine(alkyl)” group refers to a mono-substituted amine as provided herein connected, as a substituent, via a lower alkylene group.
  • a mono-substituted amine(alkyl) may be substituted or unsubstituted.
  • a mono-substituted amine(alkyl) group can include, for example, a mono-alkylamine(alkyl) group, a mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) group, a mono-arylamine(alkyl group), a mono-C 6 -C 10 arylamine(C 1 -C 6 alkyl) group and the like.
  • Examples of mono-substituted amine(alkyl) groups include, but are not limited to, —CH 2 NH(methyl), —CH 2 NH(phenyl), —CH 2 CH 2 NH(methyl), —CH 2 CH 2 NH(phenyl) and the like.
  • di-substituted amine(alkyl) refers to a di-substituted amine as provided herein connected, as a substituent, via a lower alkylene group.
  • a di-substituted amine(alkyl) may be substituted or unsubstituted.
  • a di-substituted amine(alkyl) group can include, for example, a dialkylamine(alkyl) group, a di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) group, a di-arylamine(alkyl) group, a di-C 6 -C 10 arylamine(C 1 -C 6 alkyl) group and the like.
  • di-substituted amine(alkyl)groups include, but are not limited to, —CH 2 N(methyl) 2 , —CH 2 N(phenyl)(methyl), —NCH 2 (ethyl)(methyl), —CH 2 CH 2 N(methyl) 2 , —CH 2 CH 2 N(phenyl)(methyl), —NCH 2 CH 2 (ethyl)(methyl) and the like.
  • substituents there may be one or more substituents present.
  • haloalkyl may include one or more of the same or different halogens.
  • C 1 -C 3 alkoxyphenyl may include one or more of the same or different alkoxy groups containing one, two or three atoms.
  • a radical indicates species with a single, unpaired electron such that the species containing the radical can be covalently bonded to another species.
  • a radical is not necessarily a free radical. Rather, a radical indicates a specific portion of a larger molecule.
  • the term “radical” can be used interchangeably with the term “group.”
  • pharmaceutically acceptable salt refers to a salt of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound.
  • the salt is an acid addition salt of the compound.
  • Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), a sulfuric acid, a nitric acid and a phosphoric acid (such as 2,3-dihydroxypropyl dihydrogen phosphate).
  • Pharmaceutical salts can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example formic, acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluensulfonic, trifluoroacetic, benzoic, salicylic, 2-oxopentanedioic or naphthalenesulfonic acid.
  • an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids
  • Pharmaceutical salts can also be obtained by reacting a compound with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium, a potassium or a lithium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of a carbonate, a salt of a bicarbonate, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C 1 -C 7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine and salts with amino acids such as arginine and lysine.
  • a salt such as an ammonium salt, an alkali metal salt, such as a sodium, a potassium or a lithium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of a carbonate, a salt of a bicarbonate, a salt of organic bases such as
  • a salt is formed by protonation of a nitrogen-based group (for example, NH 2 )
  • the nitrogen-based group can be associated with a positive charge (for example, NH 2 can become NH 3 + ) and the positive charge can be balanced by a negatively charged counterion (such as Cl ⁇ ).
  • HIV latency reversing agent refers to an agent (including small molecules and proteins) that stimulates HIV transcription, converting latently infected HIV cells into cells expressing replicating HIV.
  • HIV latency reversing agents include, but are not limited to protein kinase C agonists (such as prostratin, bryostatin-1 and ingenol), PD-1 inhibitors (such as nivolumab, pembrolizumab, BGB-A317, pidilizumab, AMP-224, AMP-514, PDR001, REGN2810 and MEDIO680), PD-L1 inhibitors (such as atezolizumab, durvalumab, avelumab and BMS-936559), HDAC inhibitors (such as vorinostat, panobinostat, romidepsin and valproic acid), phorbol esters (such as phorbol 12-myristate-13-acetate and (S)-tert-butyl-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][
  • Bcl protein inhibitor refers to an agent (including small molecules and proteins) that inhibit the binding of an anti-apoptic Bcl protein (such as Bcl-2, Bcl-X L , Bcl-W, Mcl-1 and Bcl-2A1) to a pro-apoptotic Bcl protein (such as Bak, Bax, Bim, Bid, tBid, Bad, Bik, PUMA, Bnip-1, Hrk, Bmf and Noxa).
  • Bcl protein inhibitors include, but are not limited to venetoclax, navitoclax, obatoclax, S55746, APG-2575, ABT-737, AMG176, AZD5991 and APG-1252.
  • Additional Bcl protein inhibitors include, but are not limited to, compounds disclosed in PCT Application Publication Nos. WO2017/132474, WO 2014/113413 and WO 2013/110890, U.S. Patent Application Publication No. 2015/0051189 and Chinese Patent Application No. CN 106565607, which are each incorporated herein by reference for the limited purpose of disclosing additional Bcl protein inhibitors.
  • FRET fluorescence resonance energy transfer
  • SPR surface plasmon resonance
  • fluorescence polarization/anisotropy fluorescence polarization/anisotropy.
  • each center may independently be of R-configuration or S-configuration or a mixture thereof.
  • the compounds provided herein may be enantiomerically pure, enantiomerically enriched, racemic mixture, diastereomerically pure, diastereomerically enriched or a stereoisomeric mixture.
  • each double bond may independently be E or Z a mixture thereof.
  • all tautomeric forms are also intended to be included.
  • valencies are to be filled with hydrogens or isotopes thereof, e.g., hydrogen-1 (protium) and hydrogen-2 (deuterium).
  • each chemical element as represented in a compound structure may include any isotope of said element.
  • a hydrogen atom may be explicitly disclosed or understood to be present in the compound.
  • the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen-1 (protium) and hydrogen-2 (deuterium).
  • reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.
  • the methods and combinations described herein include crystalline forms (also known as polymorphs, which include the different crystal packing arrangements of the same elemental composition of a compound), amorphous phases, salts, solvates and hydrates.
  • the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, ethanol or the like.
  • the compounds described herein exist in unsolvated form.
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol or the like. Hydrates are formed when the solvent is water or alcoholates are formed when the solvent is alcohol.
  • the compounds provided herein can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.
  • the term “comprising” is to be interpreted synonymously with the phrases “having at least” or “including at least”.
  • the term “comprising” means that the compound, composition or device includes at least the recited features or components, but may also include additional features or components.
  • Some embodiments disclosed herein relate to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, having the structure:
  • R 1 can be selected from hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine; each R 2 can be independently selected from halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; R 3 can be selected from hydrogen, halogen, X—R 3A ,
  • R 3A can be a substituted or unsubstituted 5 to 10 membered heteroaryl
  • R 4 can be selected from NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl
  • R 5 can be selected from —X 1 -(Alk 1 ) n -R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9
  • Alk 1 and Alk 2 can be independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl
  • R 6 can be selected from a substituted or unsubstituted C 1
  • R 1 can be halogen, for example, fluoro, chloro, bromo or iodo. In some embodiments, R 1 can be fluoro. In some embodiments, R 1 can be chloro. In some embodiments, R 1 can be hydrogen.
  • R 1 can be a substituted or unsubstituted C 1 -C 6 alkyl.
  • R 1 can be a substituted C 1 -C 6 alkyl.
  • R 1 can be an unsubstituted C 1 -C 6 alkyl.
  • suitable C 1 -C 6 alkyl groups include, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained) and hexyl (branched and straight-chained).
  • R 1 can be an unsubstituted methyl or an unsubstituted ethyl.
  • R 1 can be a substituted or unsubstituted C 1 -C 6 haloalkyl, for example, a substituted or unsubstituted mono-halo C 1 -C 6 alkyl, a substituted or unsubstituted di-halo C 1 -C 6 alkyl, a substituted or unsubstituted tri-halo C 1 -C 6 alkyl, a substituted or unsubstituted tetra-halo C 1 -C 6 alkyl or a substituted or unsubstituted penta-halo C 1 -C 6 alkyl.
  • R 1 can be an unsubstituted —CHF 2 , —CF 3 , —CH 2 CF 3 or —CF 2 CH 3 .
  • R 1 can be a substituted or unsubstituted monocyclic or bicyclic C 3 -C 6 cycloalkyl.
  • R 1 can be a substituted monocyclic C 3 -C 6 cycloalkyl.
  • R 1 can be an unsubstituted monocyclic C 3 -C 6 cycloalkyl.
  • suitable monocyclic or bicyclic C 3 -C 6 cycloalkyl groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, [1.1.1]bicyclopentyl and cyclohexyl.
  • R 1 can be a substituted or unsubstituted C 1 -C 6 alkoxy.
  • R 1 can be a substituted C 1 -C 6 alkoxy.
  • R 1 can be an unsubstituted C 1 -C 6 alkoxy.
  • suitable C 1 -C 6 alkoxy groups include, but are not limited to methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy (branched and straight-chained) and hexoxy (branched and straight-chained).
  • R 1 can be an unsubstituted methoxy or an unsubstituted ethoxy.
  • R 1 can be an unsubstituted mono-C 1 -C 6 alkylamine, for example, methylamine, ethylamine, n-propylamine, isopropylamine, n-butylamine, isobutylamine, tert-butylamine, pentylamine (branched and straight-chained) and hexylamine (branched and straight-chained).
  • R 1 can be methylamine or ethylamine.
  • R 1 can be an unsubstituted di-C 1 -C 6 alkylamine.
  • each C 1 -C 6 alkyl in the di-C 1 -C 6 alkylamine is the same. In other embodiments, each C 1 -C 6 alkyl in the di-C 1 -C 6 alkylamine is different.
  • suitable di-C 1 -C 6 alkylamine groups include, but are not limited to di-methylamine, di-ethylamine, (methyl)(ethyl)amine, (methyl)(isopropyl)amine and (ethyl)(isopropyl)amine.
  • m can be 0. When m is 0, those skilled in the art understand that the ring to which R 2 is attached is unsubstituted. In some embodiments, m can be 1. In some embodiments, m can be 2. In some embodiments, m can be 3.
  • one R 2 can be an unsubstituted C 1 -C 6 alkyl (for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl (branched and straight-chained) and hexyl (branched and straight-chained)) and any other R 2 , if present, can be independently selected from halogen (for example, fluoro or chloro), a substituted or unsubstituted C 1 -C 6 alkyl (such as those described herein), a substituted or unsubstituted C 1 -C 6 haloalkyl (such as those described herein) and a substituted or unsubstituted monocyclic or bicyclic C 3 -C 6 cycloalkyl (such as those described herein).
  • each R 2 can be independently selected from an unsubstituted C 1 -C 6 alkyl (
  • m can be 2; and each R 2 can be geminal. In some embodiments, m can be 2; and each R 2 can be vicinal. In some embodiments, m can be 2; and each R 2 can be an unsubstituted methyl. In some embodiments, m can be 2; and each R 2 can be a geminal unsubstituted methyl.
  • two R 2 groups can be taken together with the atom(s) to which they are attached to form a substituted or unsubstituted monocyclic C 3 -C 6 cycloalkyl.
  • two R 2 groups can be taken together with the atom(s) to which they are attached to form a substituted monocyclic C 3 -C 6 cycloalkyl, such as those described herein.
  • two R 2 groups can be taken together with the atom(s) to which they are attached to form an unsubstituted monocyclic C 3 -C 6 cycloalkyl, such as those described herein.
  • two R 2 groups can be taken together with the atom to which they are attached to form an unsubstituted cyclopropyl.
  • two R 2 groups can be taken together with the atom(s) to which they are attached to form a substituted or unsubstituted monocyclic 3 to 6 membered heterocyclyl.
  • two R 2 groups can be taken together with the atom(s) to which they are attached to form a substituted monocyclic 3 to 6 membered heterocyclyl.
  • two R 2 groups can be taken together with the atom(s) to which they are attached to form an unsubstituted monocyclic 3 to 6 membered monocyclic heterocyclyl.
  • the substituted monocyclic 3 to 6 membered heterocyclyl can be substituted on one or more nitrogen atoms.
  • Suitable substituted or unsubstituted monocyclic 3 to 6 membered heterocyclyl groups include, but are not limited to azidirine, oxirane, azetidine, oxetane, pyrrolidine, tetrahydrofuran, imidazoline, pyrazolidine, piperidine, tetrahydropyran, piperazine, morpholine, thiomorpholine and dioxane.
  • R 3 can be
  • R 3 can be
  • R 3 can be X—R 3A .
  • X can be —O—.
  • X can be —S—.
  • X can be —NH—.
  • R 3A can be
  • R 3A can be any organic compound
  • R 3A can be a substituted or unsubstituted 5 to 10 membered heteroaryl. In some embodiments, R 3A can be a substituted 5 to 10 membered monocyclic heteroaryl. In other embodiments, R 3A can be a substituted 5 to 10 membered bicyclic heteroaryl. In some embodiments, R 3A can be an unsubstituted 5 to 10 membered monocyclic heteroaryl. In other embodiments, R 3A can be an unsubstituted 5 to 10 membered bicyclic heteroaryl.
  • Suitable substituted or unsubstituted monocyclic or bicyclic 5 to 10 membered heteroaryl groups include, but are not limited to pyrrole, furan, thiophene, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, triazole, pyridine, pyridazine, pyrimidine, pyrazine, pyrrolo-pyrroles, pyrrolo-furans, pyrrolo-thiophenes, indole, isoindole, indolizine, indazole, benzimidazole, azaindoles, azaindazoles, purine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, quinoline, isoquinoline, quinoxaline, phthalazine, quinazoline, cinnoline, 1,8-naphthy
  • R 3 can be hydrogen. In some embodiments, R 3 can be halogen. In some embodiments, R 3 can be fluoro or chloro.
  • R 4 can be NO 2 . In some embodiments, R 4 can be cyano. In some embodiments, R 4 can be halogen.
  • R 4 can be an unsubstituted C 1 -C 6 haloalkyl, such as those described herein. In some embodiments, R 4 can be —CF 3 .
  • R 4 can be S(O)R 6 . In some embodiments, R 4 can be SO 2 R 6 . In some embodiments, R 4 can be SO 2 CF 3 .
  • R 6 can be a substituted or unsubstituted C 1 -C 6 alkyl.
  • R 6 can be a substituted C 1 -C 6 alkyl, such as those described herein.
  • R 6 can be an unsubstituted C 1 -C 6 alkyl, such as those described herein.
  • R 6 can be a substituted or unsubstituted monocyclic or bicyclic C 3 -C 6 cycloalkyl.
  • R 6 can be a substituted monocyclic or bicyclic C 3 -C 6 cycloalkyl.
  • R 6 can be an unsubstituted monocyclic or bicyclic C 3 -C 6 cycloalkyl.
  • suitable monocyclic or bicyclic C 3 -C 6 cycloalkyl groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, [1.1.1]bicyclopentyl and cyclohexyl.
  • R 6 can be a substituted or unsubstituted C 1 -C 6 haloalkyl, such as those described herein. In some embodiments, R 6 can be —CF 3 .
  • R 8 can be —X 1 -(Alk 1 ) n —R 7 .
  • X 1 can be —O—.
  • X 1 can be —S—.
  • X 1 can be —NH—.
  • Alk 1 can be unsubstituted —(CH 2 ) 1-4 —* for which “*” represents the point of attachment to R 7 . In some embodiments, Alk 1 can be
  • Alk 1 can be a substituted
  • Alk 1 can be a substituted methylene, a substituted ethylene, a substituted propylene or a substituted butylene.
  • Alk 1 can be mono-substituted, di-substituted or tri-substituted.
  • Alk 1 can be mono-substituted with a halogen (such as fluoro or chloro) or unsubstituted C 1 -C 3 alkyl, such as those described herein.
  • Alk 1 can be mono-substituted unsubstituted C 1 -C 3 haloalkyl, such as those described herein. In some embodiments, Alk 1 can be mono-substituted with fluoro or unsubstituted methyl. In some embodiments, Alk 1 can be di-substituted with one fluoro and one unsubstituted C 1 -C 3 alkyl, such as those described herein. In other embodiments, Alk 1 can be di-substituted with one unsubstituted C 1 -C 3 haloalkyl, such as those described herein, and one unsubstituted C 1 -C 3 alkyl, such as those described herein.
  • Alk 1 can be di-substituted with one fluoro and one unsubstituted methyl. In some embodiments, Alk 1 can be di-substituted with two independently selected unsubstituted C 1 -C 3 alkyl groups, such as those described herein. In some embodiments, Alk 1 can be di-substituted with unsubstituted methyl.
  • Alk 1 can be selected from:
  • n can be 0. When n is 0, those skilled in the art understand that X 1 is directly connected to R 7 . In some embodiments, n can be 1.
  • R 7 can be a substituted or unsubstituted mono-substituted amine group.
  • R 7 can be an amino group mono-substituted with a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 2 -C 6 alkenyl, a substituted or unsubstituted C 2 -C 6 alkynyl, a substituted or unsubstituted monocyclic or bicyclic C 3 -C 6 cycloalkyl, a substituted or unsubstituted monocyclic or bicyclic C 6 -C 10 aryl, a substituted or unsubstituted monocyclic or bicyclic 5 to 10 membered heteroaryl, a substituted or unsubstituted monocyclic or bicyclic 3 to 10 membered heterocyclyl, a substituted or unsubstituted monocyclic or bicyclic C 3 -C 6
  • Suitable mono-substituted amine groups include, but are not limited to —NH(methyl), —NH(isopropyl), —NH(cyclopropyl), —NH(phenyl), —NH(benzyl) and —NH(pyridine-3-yl).
  • R 7 can be a substituted or unsubstituted di-substituted amine group.
  • R 7 can be an amino group substituted with two substituents independently selected from a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 2 -C 6 alkenyl, a substituted or unsubstituted C 2 -C 6 alkynyl, a substituted or unsubstituted monocyclic or bicyclic C 3 -C 6 cycloalkyl, a substituted or unsubstituted monocyclic or bicyclic C 6 -C 10 aryl, a substituted or unsubstituted monocyclic or bicyclic 5 to 10 membered heteroaryl, a substituted or unsubstituted monocyclic or bicyclic 3 to 10 membered heterocyclyl, a substituted or unsubstituted monocyclic or bicyclic C 3 -
  • the two substituents can be the same. In other embodiments the two substituents can be different.
  • suitable di-substituted amine groups include, but are not limited to, —N(methyl) 2 , —N(ethyl) 2 , —N(isopropyl) 2 , —N(benzyl) 2 , —N(ethyl)(methyl), —N(isopropyl)(methyl), —N(ethyl)(isopropyl), —N(phenyl)(methyl) and —N(benzyl)(methyl).
  • R 7 can be selected from a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido.
  • R 7 can be a substituted or unsubstituted C 3 -C 10 cycloalkyl. In some embodiments, R 7 can be a substituted or unsubstituted monocyclic C 3 -C 10 cycloalkyl. In other embodiments, R 7 can be a substituted or unsubstituted bicyclic C 3 -C 10 cycloalkyl, for example, a bridged, fused or spiro C 3 -C 10 cycloalkyl.
  • Suitable substituted or unsubstituted monocyclic or bicyclic C 3 -C 10 cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, spiro[3.3]heptyl, spiro[2.3]hexyl, spiro[3.4]octyl, spiro[3.5]nonyl, spiro[3.6]decyl, spiro[2.4]heptyl, spiro[4.4]nonyl, spiro[4.5]decyl, spiro[2.5]octyl, spiro[3.5]nonyl, bicyclo[1.1.1]pentyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]h
  • R 7 can be a substituted or unsubstituted C 6 -C 10 spirocycloalkyl. In some embodiments, R 7 can be a substituted C 6 -C 10 spirocycloalkyl. In other embodiments, R 7 can be an unsubstituted C 6 -C 10 spirocycloalkyl.
  • R 7 can be a substituted or unsubstituted-cyclopropyl-cyclobutyl spiroalkyl, -cyclopropyl-cyclopentyl spiroalkyl, -cyclopropyl-cyclohexyl spiroalkyl, -cyclopropyl-cycloheptyl spiroalkyl, -cyclopropyl-cyclooctyl spiroalkyl, -cyclobutyl-cyclopropyl spiroalkyl, -cyclobutyl-cyclobutyl spiroalkyl, -cyclobutyl-cyclopentyl spiroalkyl, -cyclobutyl-cyclohexyl spiroalkyl, -cyclobutyl-cycloheptyl spiroalkyl, -cyclopentyl-cyclopropyl spiroalkyl
  • R 7 can be a substituted or unsubstituted 3 to 10 membered heterocyclyl. In some embodiments, R 7 can be a substituted 3 to 10 membered heterocyclyl. In other embodiments, R 7 can be an unsubstituted 3 to 10 membered heterocyclyl. In some embodiments, R 7 can be a substituted or unsubstituted monocyclic 3 to 10 membered heterocyclyl. In other embodiments, R 7 can be a substituted or unsubstituted bicyclic 5 to 10 membered heterocyclyl, for example, a fused, bridged or spiro 5 to 10 membered heterocyclyl.
  • Suitable substituted or unsubstituted 3 to 10 membered heterocyclyl groups include, but are not limited to, azidirine, oxirane, azetidine, oxetane, pyrrolidine, tetrahydrofuran, imidazoline, pyrazolidine, piperidine, tetrahydropyran, piperazine, morpholine, thiomorpholine, dioxane, 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-azaspiro[3.4]octane, 6-oxaspiro[3.4]octane, 6-oxa-2-azaspiro[3.4]octane, 7-oxa-2-azaspiro[3.5]nonane, 7-oxaspiro[3.5
  • the substituted or unsubstituted monocyclic or bicyclic 3 to 10 membered heterocyclyl can be connected to the rest of the molecule through a nitrogen atom. In other embodiments, the substituted or unsubstituted monocyclic or bicyclic 3 to 10 membered heterocyclyl can be connected to the rest of the molecule through a carbon atom. In some embodiments, the substituted monocyclic or bicyclic 3 to 10 membered heterocyclyl can be substituted on one or more nitrogen atoms.
  • R 7 can be a substituted or unsubstituted 6 to 10 membered spiro heterocyclyl. In some embodiments, R 7 can be a substituted 6 to 10 membered spiro heterocyclyl. In other embodiments, R 7 can be an unsubstituted 6 to 10 membered spiro heterocyclyl.
  • R 7 can be a substituted or unsubstituted azaspirohexane, azaspiroheptane, azaspirooctane, oxaspirohexane, oxaspiroheptane, oxaspirooctane, diazaspirohexane, diazaspiroheptane, diazaspirooctane, dioxaspirohexane, dioxaspiroheptane, dioxaspirooctane, oxa-azaspirohexane, oxa-azaspiroheptane or oxa-azaspirooctane.
  • Suitable substituted or unsubstituted 3 to 10 membered heterocyclyl groups include, but are not limited to, 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-azaspiro[3.4]octane, 6-oxaspiro[3.4]octane, 6-oxa-2-azaspiro[3.4]octane, 7-oxa-2-azaspiro[3.5]nonane, 7-oxaspiro[3.5]nonane and 2-oxa-8-azaspiro[4.5]decane.
  • the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl can be connected to the rest of the molecule through a nitrogen atom. In other embodiments, the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl can be connected to the rest of the molecule through a carbon atom. In some embodiments, the substituted 6 to 10 membered spiroheterocyclyl can be substituted on one or more nitrogen atoms.
  • R 7 can be hydroxy or amino.
  • R 7 can be unsubstituted. In other embodiments, R 7 can be substituted. In some embodiments, R 7 can be substituted with 1 or 2 substituents independently selected from an unsubstituted C 1 -C 6 alkyl (such as those described herein), an unsubstituted C 1 -C 6 alkoxy (such as those described herein), fluoro, chloro, hydroxy and —SO 2 -(unsubstituted C 1 -C 6 alkyl).
  • the C 1 -C 6 alkoxy, C 3 -C 10 cycloalkyl, 3 to 10 membered heterocyclyl, mono-substituted amine group, di-substituted amine group, N-carbamyl, C-amido and N-amido groups of R 7 can be substituted with 1 or 2 substituents independently selected from any of the aforementioned substituents.
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 7 can be
  • R 5 can be —X 2 —(CHR 8 )-(Alk 2 ) p -X 3 —R 9 .
  • X 2 can be —O—.
  • X 2 can be —S—.
  • X 2 can be —NH—.
  • X 3 can be —O—.
  • X 3 can be —S—.
  • X 3 can be —NH—.
  • X 2 can be —NH— and X 3 can be —S—.
  • X 2 can be —O— and X 3 can be —S—.
  • X 2 can be —NH— and X 3 can be —O— and X 3 can be —S—.
  • X 2 can be —NH— and X 3 can be —O—.
  • X 2 can be —O— and X 3 can be —O—.
  • Alk 2 can be unsubstituted —(CH 2 ) 1-4 —* for which “*” represents the point of attachment to X 3 .
  • Alk 2 can be an unsubstituted methylene, an unsubstituted ethylene, an unsubstituted propylene or an unsubstituted butylene.
  • Alk 2 can be
  • Alk 2 can be a substituted
  • Alk 2 can be a substituted methylene, a substituted ethylene, a substituted propylene or a substituted butylene. In some embodiments, Alk 2 can be mono-substituted, di-substituted or tri-substituted. In some embodiments, Alk 2 can be mono-substituted with fluoro or unsubstituted C 1 -C 3 alkyl, such as those described herein. In some embodiments, Alk 2 can be mono-substituted with fluoro or unsubstituted methyl.
  • Alk 2 can be di-substituted with one fluoro and one unsubstituted C 1 -C 3 alkyl, such as those described herein. In some embodiments, Alk 2 can be di-substituted with one fluoro and one unsubstituted methyl. In some embodiments, Alk 2 can be di-substituted with two independently selected unsubstituted C 1 -C 3 alkyl, such as those described herein. In some embodiments, Alk 2 can be di-substituted with unsubstituted methyl.
  • Alk 2 can be selected from:
  • p can be 0. When p is 0, those skilled in the art understand that the (CHR 8 ) group is directly connected to X 3 . In some embodiments, p can be 1.
  • the C 1 -C 6 alkyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) of R 8 can be a substituted or unsubstituted C 1 -C 6 alkyl such as those described herein.
  • the 3 to 10 membered heterocyclyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) of R 8 can be monocyclic.
  • the 3 to 10 membered heterocyclyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) can be bicyclic.
  • the 3 to 10 membered heterocyclyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) can be connected to the C 1 -C 6 alkyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) through a carbon atom.
  • the 3 to 10 membered heterocyclyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) can be unsubstituted.
  • the 3 to 10 membered heterocyclyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) can be substituted. In some embodiments, the 3 to 10 membered heterocyclyl of the substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl) can be substituted on one or more nitrogen atoms.
  • Suitable substituted or unsubstituted monocyclic or bicyclic 3 to 10 membered heterocyclyl groups of R 8 include, but are not limited to azidirine, oxirane, azetidine, oxetane, pyrrolidine, tetrahydrofuran, imidazoline, pyrazolidine, piperidine, tetrahydropyran, piperazine, morpholine, thiomorpholine, dioxane, 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-azaspiro[3.4]octane, 6-oxaspiro[3.4]octane, 6-oxa-2-azaspiro[3.4]octane, 7-oxa-2-azaspiro[3.5]nonane, 7
  • the C 1 -C 6 alkyl of R 8 can be an unsubstituted methyl or an unsubstituted ethyl and the substituted or unsubstituted 3 to 10 membered heterocyclyl of R 8 can be a piperidine, tetrahydropyran, piperazine, morpholine, thiomorpholine, dioxane, 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-azaspiro[3.4]octane, 6-oxaspiro[3.4]octane, 6-oxa-2-azaspiro[3.4]octane, 7-oxa-2-azaspiro[3.5]nonane, 7-oxaspiro[3.5]nonane or 2-ox
  • R 8 can be a substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl).
  • the C 1 -C 6 alkyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) of R 8 can be a substituted or unsubstituted C 1 -C 6 alkyl, such as those described herein.
  • the C 1 -C 6 alkyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be unsubstituted.
  • the 6 to 10 membered spiro heterocyclyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be connected to the C 1 -C 6 alkyl of R 8 through a nitrogen atom.
  • the 6 to 10 membered spiro heterocyclyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be connected to the C 1 -C 6 alkyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) through a carbon atom.
  • the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be unsubstituted.
  • 6 to 10 membered spiro heterocyclyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be substituted.
  • the 6 to 10 membered spiro heterocyclyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be substituted on one or more nitrogen atoms.
  • the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be an azaspirohexane, azaspiroheptane, azaspirooctane, oxaspirohexane, oxaspiroheptane, oxaspirooctane, diazaspirohexane, diazaspiroheptane, diazaspirooctane, dioxaspirohexane, dioxaspiroheptane, dioxaspirooctane, oxa-azaspirohexane, oxa-azaspiroheptane or oxa-azaspirooctane.
  • Suitable substituted or unsubstituted 6 to 10 membered spiro heterocyclyl of R 8 include, but are not limited to, 2-azaspiro[3.3]heptane, 2-oxaspiro[3.3]heptane, 2,6-diazaspiro[3.3]heptane, 2-oxa-6-azaspiro[3.3]heptane, 2-azaspiro[3.4]octane, 6-oxaspiro[3.4]octane, 6-oxa-2-azaspiro[3.4]octane, 7-oxa-2-azaspiro[3.5]nonane, 7-oxaspiro[3.5]nonane and 2-oxa-8-azaspiro[4.5]decane.
  • the C 1 -C 6 alkyl of the substituted or unsubstituted 6 to 10 membered spiro heterocyclyl(C 1 -C 6 alkyl) can be an unsubstituted methyl or an unsubstituted ethyl and the 6 to 10 membered spiro heterocyclyl of R 8 can be an azaspirohexane, azaspiroheptane, azaspirooctane, oxaspirohexane, oxaspiroheptane, oxaspirooctane, diazaspirohexane, diazaspiroheptane, diazaspirooctane, dioxaspirohexane, dioxaspiroheptane, dioxaspirooctane, oxa-azaspirohexane, oxa-azaspirohexan
  • R 8 can be a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl), for example, a di-C 1 -C 6 alkylamine(ethyl), di-C 1 -C 6 alkylamine(propyl), di-C 1 -C 6 alkylamine(butyl), di-C 1 -C 6 alkylamine(pentyl) or di-C 1 -C 6 alkylamine(hexyl).
  • each C 1 -C 6 alkyl group in the di-C 1 -C 6 alkylamine can be the same.
  • each C 1 -C 6 alkyl group in the di-C 1 -C 6 alkylamine can be different.
  • Suitable substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) include, but are not limited to, —N(methyl) 2 , —N(ethyl) 2 , —N(n-propyl) 2 , —N(isopropyl) 2 , —N(t-butyl) 2 , —N(ethyl)(methyl), —N(isopropyl)(methyl), —N(t-butyl)(methyl) and —N(isopropyl)(ethyl); each connected to a substituted or unsubstituted C 1 -C 6 alkyl group.
  • R 8 can be a substituted or unsubstituted di-methylamine(C 1 -C 6 alkyl), for example,
  • R 8 can be a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl), for example, a substituted or unsubstituted mono-C 1 -C 6 alkylamine(ethyl), mono-C 1 -C 6 alkylamine(propyl), mono-C 1 -C 6 alkylamine(butyl), mono-C 1 -C 6 alkylamine(pentyl) or mono-C 1 -C 6 alkylamine(hexyl).
  • the C 1 -C 6 alkyl of the unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) group can be an unsubstituted C 1 -C 6 alkyl, such as those described herein.
  • R 8 can be unsubstituted. In other embodiments, R 8 can be substituted. In some embodiments, R 8 can be substituted with 1 or 2 substituents independently selected from an unsubstituted C 1 -C 6 alkyl (such as those described herein), an unsubstituted C 1 -C 6 alkoxy (such as those described herein), an unsubstituted di-C 1 -C 6 alkylamine (such as those described herein), an unsubstituted acyl(C 1 -C 6 alkyl) (for example, acetyl or benzoyl), an unsubstituted C-carboxy (for example, —CO 2 H, —CO 2 —C 1 -C 6 alkyl, —CO 2 —C 3 -C 6 cycloalkyl or —CO 2 —C 6 -C 10 aryl), fluoro, chloro and hydroxy.
  • R 8 can be substituted with 1 or 2 substituents independently
  • the 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and mono-C 1 —C 6 alkylamine(C 1 -C 6 alkyl) groups of R 8 can be substituted with 1 or 2 substituents independently selected from any of the aforementioned substituents.
  • R 8 can be:
  • R can be any organic radical
  • R 9 can be a substituted or unsubstituted monocyclic or bicyclic C 6 -C 10 aryl. In some embodiments, R 9 can be a substituted monocyclic or bicyclic C 6 -C 10 aryl. In other embodiments, R 9 can be an unsubstituted monocyclic or bicyclic C 6 -C 10 aryl. In some embodiments, R 9 can be a substituted phenyl or a substituted naphthyl. In some embodiments, R 9 can be an unsubstituted phenyl or an unsubstituted naphthyl.
  • R 9 can be a substituted or unsubstituted 5 to 10 membered heteroaryl. In some embodiments, R 9 can be a substituted 5 to 10 membered heteroaryl. In other embodiments, R 9 can be an unsubstituted 5 to 10 membered heteroaryl. In some embodiments, R 9 can be a monocyclic substituted or unsubstituted 5 to 10 membered heteroaryl. In other embodiments, R 9 can be a bicyclic substituted or unsubstituted 5 to 10 membered heteroaryl.
  • Suitable substituted or unsubstituted monocyclic or bicyclic 5 to 10 membered heteroaryl include, but are not limited to, pyrrole, furan, thiophene, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, triazole, pyridine, pyridazine, pyrimidine, pyrazine, pyrrolo-pyrroles, pyrrolo-furans, pyrrolo-thiophenes, indole, isoindole, indolizine, indazole, benzimidazole, azaindoles, purine, benzofuran, isobenzofuran, benzothiophene, isobenzothiophene, quinoline, isoquinoline, quinoxaline, phthalazine, quinazoline, cinnoline, 1,8-naphthyridine, pyrid
  • R 3 is hydrogen or halogen.
  • an embodiment provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine;
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; or
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl, or two R 2 groups taken together with the atom(s) to which they are attached form a substituted or unsubstituted C 3 -C 6 cycloalkyl or a substituted or unsubstituted 3 to 6 membered heterocyclyl;
  • R 3 is hydrogen or halogen
  • R 4 is selected from the group consisting of NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl;
  • R 5 is selected from the group consisting of —X 1 -(Alk 1 ) n —R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9 ;
  • Alk 1 and Alk 2 are independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl;
  • R 6 is selected from the group consisting of a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl;
  • R 7 is selected from a substituted or unsubstituted C 1 -C 6 alkoxy, a substituted or unsubstituted C 3 -C 10 cycloalkyl, a substituted or unsubstituted 3 to 10 membered heterocyclyl, hydroxy, amino, a substituted or unsubstituted mono-substituted amine group, a substituted or unsubstituted di-substituted amine group, a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido;
  • R 8 is selected from a substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl);
  • R 9 is selected from a substituted or unsubstituted 5 to 10 membered heteroaryl and a substituted or unsubstituted C 6 -C 10 aryl;
  • n 0, 1, 2 or 3;
  • n and p are independently selected from 0 and 1;
  • X 1 , X 2 and X 3 are independently selected from the group consisting of —O—, —S— and —NH—.
  • R 3 is selected from the group consisting of X—R 3A ,
  • an embodiment provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine;
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; or
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl, or two R 2 groups taken together with the atom(s) to which they are attached form a substituted or unsubstituted C 3 -C 6 cycloalkyl or a substituted or unsubstituted 3 to 6 membered heterocyclyl;
  • R 3 is selected from the group consisting of X—R 3A ,
  • R 3A is a substituted or unsubstituted 5 to 10 membered heteroaryl
  • R 4 is selected from the group consisting of NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl;
  • R 5 is selected from the group consisting of —X 1 -(Alk 1 ) n —R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9 ;
  • Alk 1 and Alk 2 are independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl;
  • R 6 is selected from the group consisting of a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl;
  • R 7 is selected from a substituted or unsubstituted C 1 -C 6 alkoxy, a substituted or unsubstituted C 3 -C 10 cycloalkyl, a substituted or unsubstituted 3 to 10 membered heterocyclyl, hydroxy, amino, a substituted or unsubstituted mono-substituted amine group, a substituted or unsubstituted di-substituted amine group, a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido;
  • R 8 is selected from a substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl);
  • R 9 is selected from a substituted or unsubstituted 5 to 10 membered heteroaryl and a substituted or unsubstituted C 6 -C 10 aryl;
  • n 0, 1, 2 or 3;
  • n and p are independently selected from 0 and 1;
  • X, X 1 , X 2 and X 3 are independently selected from the group consisting of —O—, —S— and —NH—.
  • R 3 is selected from the group consisting of X—R 3A ,
  • an embodiment provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine;
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; or
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl, or two R 2 groups taken together with the atom(s) to which they are attached form a substituted or unsubstituted C 3 -C 6 cycloalkyl or a substituted or unsubstituted 3 to 6 membered heterocyclyl;
  • R 3 is selected from the group consisting of X—R 3A ,
  • R 3A is a substituted or unsubstituted 5 to 10 membered heteroaryl
  • R 4 is selected from the group consisting of NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl;
  • R 5 is selected from the group consisting of —X 1 -(Alk 1 ) n —R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9 ;
  • Alk 1 and Alk 2 are independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl;
  • R 6 is selected from the group consisting of a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl;
  • R 7 is selected from a substituted or unsubstituted C 1 -C 6 alkoxy, a substituted or unsubstituted C 3 -C 10 cycloalkyl, a substituted or unsubstituted 3 to 10 membered heterocyclyl, hydroxy, amino, a substituted or unsubstituted mono-substituted amine group, a substituted or unsubstituted di-substituted amine group, a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido;
  • R 8 is selected from a substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl);
  • R 9 is selected from a substituted or unsubstituted 5 to 10 membered heteroaryl and a substituted or unsubstituted C 6 -C 10 aryl;
  • n 0, 1, 2 or 3;
  • n and p are independently selected from 0 and 1;
  • X 1 and X 2 are —NH—;
  • X and X 3 are independently selected from the group consisting of —O—, —S— and —NH—.
  • R 1 is as described above with the proviso that it is not —CH 2 F, —CHF 2 or —CF 3 ;
  • R 3 is selected from the group consisting of X—R 3A ,
  • an embodiment provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine, with the proviso that R 1 is not —CH 2 F, —CHF 2 or —CF 3 ;
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; or
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl, or two R 2 groups taken together with the atom(s) to which they are attached form a substituted or unsubstituted C 3 -C 6 cycloalkyl or a substituted or unsubstituted 3 to 6 membered heterocyclyl;
  • R 3 is selected from the group consisting of X—R 3A ,
  • R 3A is a substituted or unsubstituted 5 to 10 membered heteroaryl
  • R 4 is selected from the group consisting of NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl;
  • R 5 is selected from the group consisting of —X 1 -(Alk 1 ) n —R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9 ;
  • Alk 1 and Alk 2 are independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl;
  • R 6 is selected from the group consisting of a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl;
  • R 7 is selected from a substituted or unsubstituted C 1 -C 6 alkoxy, a substituted or unsubstituted C 3 -C 10 cycloalkyl, a substituted or unsubstituted 3 to 10 membered heterocyclyl, hydroxy, amino, a substituted or unsubstituted mono-substituted amine group, a substituted or unsubstituted di-substituted amine group, a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido;
  • R 8 is selected from a substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl);
  • R 9 is selected from a substituted or unsubstituted 5 to 10 membered heteroaryl and a substituted or unsubstituted C 6 -C 10 aryl;
  • n 0, 1, 2 or 3;
  • n and p are independently selected from 0 and 1;
  • X 1 and X 2 are —NH—;
  • X and X 3 are independently selected from the group consisting of —O—, —S— and —NH—.
  • R 1 is —CH 2 F, —CHF 2 or —CF 3 ;
  • R 3 is selected from the group consisting of X—R 3A ,
  • an embodiment provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is —CH 2 F, —CHF 2 or —CF 3 ;
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; or
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl, or two R 2 groups taken together with the atom(s) to which they are attached form a substituted or unsubstituted C 3 -C 6 cycloalkyl or a substituted or unsubstituted 3 to 6 membered heterocyclyl;
  • R 3 is selected from the group consisting of X—R 3A ,
  • R 3A is a substituted or unsubstituted 5 to 10 membered heteroaryl
  • R 4 is selected from the group consisting of NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl;
  • R 5 is selected from the group consisting of —X 1 -(Alk 1 ) n —R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9 ;
  • Alk 1 and Alk 2 are independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl;
  • R 6 is selected from the group consisting of a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl;
  • R 7 is selected from a substituted or unsubstituted C 1 -C 6 alkoxy, a substituted or unsubstituted C 3 -C 10 cycloalkyl, a substituted or unsubstituted 3 to 10 membered heterocyclyl, hydroxy, amino, a substituted or unsubstituted mono-substituted amine group, a substituted or unsubstituted di-substituted amine group, a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido;
  • R 8 is selected from a substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl);
  • R 9 is selected from a substituted or unsubstituted 5 to 10 membered heteroaryl and a substituted or unsubstituted C 6 -C 10 aryl;
  • n 0, 1, 2 or 3;
  • n and p are independently selected from 0 and 1;
  • X 1 and X 2 are —NH—;
  • X and X 3 are independently selected from the group consisting of —O—, —S— and —NH—.
  • R 3 is selected from the group consisting of X—R 3A ,
  • an embodiment provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine;
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl; or
  • each R 2 is independently selected from the group consisting of halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl, or two R 2 groups taken together with the atom(s) to which they are attached form a substituted or unsubstituted C 3 -C 6 cycloalkyl or a substituted or unsubstituted 3 to 6 membered heterocyclyl;
  • R 3 is selected from the group consisting of X—R 3A ,
  • R 3A is a substituted or unsubstituted 5 to 10 membered heteroaryl
  • R 4 is selected from the group consisting of NO 2 , S(O)R 6 , SO 2 R 6 , halogen, cyano and an unsubstituted C 1 -C 6 haloalkyl;
  • R 5 is selected from the group consisting of —X-(Alk 1 ) n —R 7 and —X 2 (CHR 8 )-(Alk 2 ) p -X 3 —R 9 ;
  • Alk 1 and Alk 2 are independently selected from an unsubstituted C 1 -C 4 alkylene and a C 1 -C 4 alkylene substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, an unsubstituted C 1 -C 3 alkyl and an unsubstituted C 1 -C 3 haloalkyl;
  • R 6 is selected from the group consisting of a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl and a substituted or unsubstituted C 3 -C 6 cycloalkyl;
  • R 7 is selected from a substituted or unsubstituted C 1 -C 6 alkoxy, a substituted or unsubstituted C 3 -C 10 cycloalkyl, a substituted or unsubstituted 3 to 10 membered heterocyclyl, hydroxy, amino, a substituted or unsubstituted mono-substituted amine group, a substituted or unsubstituted di-substituted amine group, a substituted or unsubstituted N-carbamyl, a substituted or unsubstituted C-amido and a substituted or unsubstituted N-amido;
  • R 8 is selected from a substituted or unsubstituted 3 to 10 membered heterocyclyl(C 1 -C 6 alkyl), a substituted or unsubstituted di-C 1 -C 6 alkylamine(C 1 -C 6 alkyl) and a substituted or unsubstituted mono-C 1 -C 6 alkylamine(C 1 -C 6 alkyl);
  • R 9 is selected from a substituted or unsubstituted 5 to 10 membered heteroaryl and a substituted or unsubstituted C 6 -C 10 aryl;
  • n 0, 1, 2 or 3;
  • n and p are independently selected from 0 and 1;
  • X 1 and X 2 are —O—;
  • X and X 3 are independently selected from the group consisting of —O—, —S— and —NH—.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof can be selected from a compound of Formula (Ia), Formula (Ib), Formula (Ic) and Formula (Id):
  • R 3 can be hydrogen
  • R 4 can be nitro or —SO 2 CF 3 .
  • R 1 can be fluoro, chloro, —CH 3 , —CH 2 CH 3 , —CHF 2 , —CF 3 , —CH 2 CF 3 , —CF 2 CH 3 , —OCH 3 , —OCH 2 CH 3 , —NHCH 3 , —NHCH 2 CH 3 , —N(CH 3 ) 2 or —N(CH 2 CH 3 ) 2 .
  • R 5 can be —O—R 7 or —NH—R 7 . In some embodiments Formulae (Ia), (Ib), (Ic) and/or (Id), R 5 can be —O-Alk 1 -R 7 or —NH-Alk 1 -R 7 . In some embodiments of Formulae (Ia), (Ib), (Ic) and/or (Id), Alk 1 can be an unsubstituted methylene, an unsubstituted ethylene, or an ethylene mono-substituted with —CH 3 .
  • R 7 can be an unsubstituted cyclohexanyl or a cyclohexanyl substituted with one or two substituents independently selected from hydroxy, amino, fluoro and unsubstituted C 1 -C 3 alkyl (such as those described herein).
  • R 7 can be a substituted or unsubstituted monocyclic 5 or 6 membered heterocyclyl, for example, pyrrolidine, piperidine, morpholine, piperazine or tetrahydropyran; wherein each of the aforementioned substituted groups can be substituted with 1 or 2 substituents independently selected from hydroxy, amino, fluoro, an unsubstituted C 1 -C 3 alkyl (such as those described herein), an unsubstituted C 1 -C 3 alkoxy (such as those described herein), or —SO 2 CH 3 .
  • R 7 can be connected to Alk 1 by a nitrogen atom. In some embodiments of Formulae (Ia), (Ib), (Ic) and/or (Id), R 7 can be connected to Alk 1 by a carbon atom. In some embodiments of Formulae (Ia), (b), (Ic) and/or (Id), R 7 can be substituted on one or more nitrogen atoms.
  • R 5 can be —NH—(CHR 8 )-Alk 2 -S—R 9 , —O—(CHR 8 )-Alk 2 -S—R 9 , —NH—(CHR 8 )-Alk 2 -O—R 9 or —O—(CHR 8 )-Alk 2 -O—R 9 .
  • Alk 2 can be an unsubstituted methylene, an unsubstituted ethylene, a methylene mono-substituted with —CH 3 or a methylene di-substituted with —CH 3 .
  • R 8 can be an unsubstituted di-C 1 -C 3 alkylamine(methyl) or an unsubstituted di-C 1 -C 3 alkylamine(ethyl).
  • R 9 can be a substituted or unsubstituted 5 to 7 membered heterocyclyl(C 1 -C 6 alkyl); wherein the C 1 -C 6 alkyl can be am unsubstituted methyl, an unsubstituted ethyl or an unsubstituted n-propyl; the 5 to 7 membered heterocyclyl can (a) be monocyclic or spiro, (b) include 1 oxygen atom, 1 nitrogen atom, or 1 oxygen atom and one nitrogen atom, (c) be unsubstituted or substituted with 1 or 2 substituents independently selected from an unsubstituted C 1 -C 3 alkyl (such as those described herein), —N(CH 3 ) 2 , —N(CH 2 CH 3 ) 2 , an unsubstituted acetyl, —CO 2 H, fluor
  • Examples of a compound of Formula (I) include:
  • FIG. 3 provides the chemical names and structures for examples of the compounds of Formula (I) listed above in which R 3 is hydrogen or halogen, along with other examples of such compounds.
  • the compound of Formula (I) is a compound selected from FIG. 3 , or a pharmaceutically acceptable salt of any of the compounds listed in FIG. 3 .
  • FIGS. 4A, 4B and 4C provide the chemical names and structures for examples of the compounds of Formula (I) listed above in which R 3 is X—R 3A ,
  • the compound of Formula (I) is a compound selected from FIGS.
  • the compound of Formula (I) is a compound selected from FIG. 3 , or a pharmaceutically acceptable salt of any of the compounds listed in FIG. 3 .
  • the compound of Formula (I) is a compound selected from FIG. 4A , or a pharmaceutically acceptable salt of any of the compounds listed in FIG. 4A .
  • the compound of Formula (I) is a compound selected from FIG. 4B , or a pharmaceutically acceptable salt of any of the compounds listed in FIG. 4B .
  • the compound of Formula (I) is a compound selected from FIG. 4C , or a pharmaceutically acceptable salt of any of the compounds listed in FIG. 4C .
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can have increased metabolic and/or plasma stability. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can be more resistant to hydrolysis and/or more resistant to enzymatic transformations. In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can have improved properties.
  • a non-limiting list of example properties include, but are not limited to, increased biological half-life, increased bioavailability, increase potency, a sustained in vivo response, increased dosing intervals, decreased dosing amounts, decreased cytotoxicity, reduction in required amounts for treating disease conditions, a reduction of morbidity or mortality in clinical outcomes, decrease in or prevention of opportunistic infections, increased subject compliance and increased compatibility with other medications.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can have more potent anticancer activity (for example, a lower EC 50 in a cell replication assay) as compared to the current standard of care (such as venetoclax).
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can have more potent antiviral activity (for example, a lower EC 50 in a HIV replicon assay) as compared to the current standard of care (such as dolutegravir).
  • the coupling reaction reactions between compounds of the general Formulae A and B to form compounds of the Formula (I) as illustrated in General Scheme 1 can be carried out in a manner similar to the reactions as described herein in the Examples, by appropriate adjustment of the reagents and conditions described in the Examples. Any preliminary reaction steps required to form starting compounds of the general Formula A and B, or other precursors, can be carried out by those skilled in the art.
  • R 1 , R 2 , R 3 R 4 , R 5 and m can be as described herein.
  • compositions that can include an effective amount of one or more compounds described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable carrier, diluent, excipient or combination thereof.
  • composition refers to a mixture of one or more compounds and/or salts disclosed herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition facilitates administration of the compound to an organism.
  • Pharmaceutical compositions can also be obtained by reacting compounds with inorganic or organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid and salicylic acid.
  • Pharmaceutical compositions will generally be tailored to the specific intended route of administration.
  • physiologically acceptable defines a carrier, diluent or excipient that does not abrogate the biological activity and properties of the compound nor cause appreciable damage or injury to an animal to which delivery of the composition is intended.
  • a “carrier” refers to a compound that facilitates the incorporation of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • a “diluent” refers to an ingredient in a pharmaceutical composition that lacks appreciable pharmacological activity but may be pharmaceutically necessary or desirable.
  • a diluent may be used to increase the bulk of a potent drug whose mass is too small for manufacture and/or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation.
  • a common form of diluent in the art is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mimics the pH and isotonicity of human blood.
  • an “excipient” refers to an essentially inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition.
  • stabilizers such as anti-oxidants and metal-chelating agents are excipients.
  • the pharmaceutical composition comprises an anti-oxidant and/or a metal-chelating agent.
  • a “diluent” is a type of excipient.
  • compositions described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or carriers, diluents, excipients or combinations thereof. Proper formulation is dependent upon the route of administration chosen. Techniques for formulation and administration of the compounds described herein are known to those skilled in the art.
  • compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. Additionally, the active ingredients are contained in an amount effective to achieve its intended purpose. Many of the compounds used in the pharmaceutical combinations disclosed herein may be provided as salts with pharmaceutically compatible counterions.
  • a compound, salt and/or composition include, but not limited to, oral, rectal, pulmonary, topical, aerosol, injection, infusion and parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, intrathecal, direct intraventricular, intraperitoneal, intranasal and intraocular injections.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can be administered orally.
  • the liposomes will be targeted to and taken up selectively by the organ. For example, intranasal or pulmonary delivery to target a respiratory disease or condition may be desirable.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • Compositions that can include a compound and/or salt described herein formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container and labeled for treatment of an indicated condition.
  • Some embodiments described herein relate to a method for treating a cancer or a tumor described herein that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) to a subject having a cancer described herein.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) to a subject having a cancer described herein.
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for treating a cancer or a tumor described herein.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for treating a cancer or a tumor described herein.
  • Some embodiments described herein relate to a method for inhibiting replication of a malignant growth or a tumor described herein that can include contacting the growth or the tumor with an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof).
  • Other embodiments described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for inhibiting replication of a malignant growth or a tumor described herein.
  • the use can include contacting the growth or the tumor with the medicament.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for inhibiting replication of a malignant growth or a tumor described herein.
  • Some embodiments described herein relate to a method for treating a cancer described herein that can include contacting a malignant growth or a tumor described herein with an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof).
  • Other embodiments described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for treating a cancer described herein.
  • the use can include contacting the malignant growth or a tumor described herein with the medicament.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for contacting a malignant growth or a tumor described herein, wherein the malignant growth or tumor is due to a cancer described herein.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • suitable malignant growths, cancers and tumors include, but are not limited to: bladder cancers, brain cancers, breast cancers, bone marrow cancers, cervical cancers, colorectal cancers, esophageal cancers, hepatocellular cancers, lymphoblastic leukemias, follicular lymphomas, lymphoid malignancies of T-cell or B-cell origin, melanomas, myelogenous leukemias, Hodgkin's lymphoma, Non-Hodgkin's lymphoma, head and neck cancers (including oral cancers), ovarian cancers, non-small cell lung cancer, chronic lymphocytic leukemias, myelomas (including multiple myelomas), prostate cancer, small cell lung cancer, spleen cancers, polycythemia vera, thyroid cancers, endometrial cancer, stomach cancers, gallbladder cancer, bile duct cancers, testicular cancers, neuroblastomas, osteosar
  • a malignant growth, cancer or tumor can become resistant to one or more anti-proliferative agents.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • anti-proliferative agents examples include, but are not limited to, Bcl-2 inhibitors (such as venetoclax, navitoclax, obatoclax, S55746, APG-1252, APG-2575 and ABT-737).
  • Bcl-2 inhibitors such as venetoclax, navitoclax, obatoclax, S55746, APG-1252, APG-2575 and ABT-737.
  • the malignant growth, cancer or tumor, that has become resistant to one or more anti-proliferative agents can be a malignant growth, cancer or tumor, described herein.
  • Some embodiments described herein relate to a method for inhibiting the activity of Bcl-2 that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) to a subject and can also include contacting a cell that expresses Bcl-2 with an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof).
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) in the manufacture of a medicament for inhibiting the activity of Bcl-2 in a subject or, in the manufacture of a medicament for inhibiting the activity of Bcl-2, wherein the use comprises contacting a cell that expresses Bcl-2.
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) for inhibiting the activity of Bcl-2 in a subject; or for inhibiting the activity of Bcl-2 by contacting a cell that expresses Bcl-2.
  • Some embodiments described herein relate to a method of ameliorating or treating a HIV infection that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof), and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof)
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for ameliorating or treating a HIV infection in a subject suffering from the HIV infection; or, in the manufacture of a medicament for ameliorating or treating a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for ameliorating or treating a HIV infection in a subject suffering from the HIV infection; or for ameliorating or treating a HIV infection by contacting a cell infected with HIV.
  • the cells can be in a subject.
  • the cells can be CD4+T cells.
  • the CD4+T cells can be in a subject.
  • Some embodiments described herein relate to a method of reducing the population of HIV infected cells that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof), and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or, for reducing the population of HIV infected cells, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or for reducing the population of HIV infected cells by contacting a cell infected with HIV.
  • the cells can be in a subject.
  • the cells can be CD4+T cells.
  • the CD4+T cells can be in a subject.
  • Some embodiments described herein relate to a method of reducing the reoccurrence of a HIV infection in a subject that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a
  • inventions described herein relate to the use of an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or, for reducing the reoccurrence of a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • an effective amount of a HIV latency reversing agent or a pharmaceutically acceptable salt thereof
  • Still other embodiments described herein relate to an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or for reducing the reoccurrence of a HIV infection by contacting a cell infected with HIV.
  • the cells can be in a subject.
  • the cells can be CD4+T cells.
  • the CD4+T cells can be in a subject.
  • Some embodiments described herein relate to a method of ameliorating or treating a HIV infection that can include administering an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • inventions described herein relate to the use of an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for ameliorating or treating a HIV infection; or, in the manufacture of a medicament for ameliorating or treating a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • the cells can be in a subject.
  • the cells can be CD4+T cells.
  • the CD4+T cells can be in a subject.
  • Some embodiments described herein relate to a method of reducing the population of HIV infected cells that can include administering an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • inventions described herein relate to the use of an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the population of HIV infected cells; or, in the manufacture of a medicament for reducing the population of HIV infected cells, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • Still other embodiments described herein relate to an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, for reducing the population of HIV infected cells in a subject suffering from the HIV infection; or for reducing the population of HIV infected cells by contacting a cell infected with HIV.
  • the cells can be in a subject.
  • the cells can be CD4+T cells.
  • the CD4+T cells can be in a subject.
  • Some embodiments described herein relate to a method of reducing the reoccurrence of a HIV infection in a subject that can include administering an effective amount of a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) or a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof to a subject suffering from the HIV infection; and can also include contacting a cell infected with HIV with an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a pharmaceutical composition that includes a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof)
  • inventions described herein relate to the use of an effective amount of a Bcl protein inhibitor, or a pharmaceutically acceptable salt thereof, and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the reoccurrence of a HIV infection in a subject suffering from the HIV infection; or, for reducing the reoccurrence of a HIV infection, wherein the use comprises contacting a cell infected with HIV with the medicament.
  • the cells can be in a subject.
  • the cells can be CD4+T cells.
  • the CD4+T cells can be in a subject.
  • the HIV latency reversing agent can be a protein kinase C agonist, a PD-1 inhibitor, a PD-L1 inhibitor, an HDAC inhibitor, a phorbol ester or a bromodomain inhibitor.
  • the HIV latency reversing agent can be a protein kinase C agonist, which includes, but is not limited to prostratin, bryostatin-1 and ingenol.
  • the HIV latency reversing agent can be a PD-1 inhibitor, which includes, but is not limited to nivolumab, pembrolizumab, BGB-A317, pidilizumab, AMP-224, AMP-514, PDR001, REGN2810 and MEDI0680.
  • the HIV latency reversing agent can be a PD-L1 inhibitor, which includes, but is not limited to atezolizumab, durvalumab, avelumab and BMS-936559.
  • the HIV latency reversing agent can be a HDAC inhibitor, which includes, but is not limited to vorinostat, panobinostat, romidepsin and valproic acid.
  • the HIV latency reversing agent can be a phorbol ester, which includes, but is not limited to phorbol 12-myristate-13-acetate and (S)-tert-butyl-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetate.
  • the HIV latency reversing agent can be a bromodomain inhibitor, which includes, but is not limited to JQ1, I-BET762, OTX015, I-BET151, CPI203, PFI-1, MS436, CPI-0610, RVX2135, FT-1101, BAY1238097, INCB054329, TEN-010, GSK2820151, ZEN003694, BAY-299, BMS-986158, ABBV-075 and GS-5829.
  • a combination of two or more HIV latency reversing agents may be used.
  • the Bcl protein inhibitor of Formula (I) can be a selective Bcl-2 inhibitor, a selective Bcl-X L inhibitor, a selective Bcl-W inhibitor, a selective Mcl-1 inhibitor or a selective Bcl-2A1 inhibitor. In some embodiments, the Bcl protein inhibitor of Formula (I) can inhibit more than one Bcl protein. In some embodiments, the Bcl protein inhibitor can be an inhibitor of the activity of Bcl-2 and one of Bcl-X L , Bcl-W, Mcl-1 and Bcl-2A1. In some embodiments, the Bcl protein inhibitor can be an inhibitor of the activity of Bcl-X L and one of Bcl-W, Mcl-1 and Bcl-2A1.
  • the Bcl protein inhibitor of Formula (I) can inhibit both Bcl-2 and Bcl-X L .
  • the Bcl protein inhibitor can be venetoclax, navitoclax, obatoclax, ABT-737, S55746, AT-101, APG-1252, APG-2575, AMG176 or AZD5991, or a combination of any of the foregoing.
  • the methods of ameliorating or treating a HIV infection can also include the use of one or more agents selected from a non-nucleoside reverse transcriptase inhibitor (NNRTI), a nucleoside reverse transcriptase inhibitor (NRTI), a protease inhibitor (PI), a fusion/entry inhibitor, an integrase strand transfer inhibitor (INSTI), a HIV vaccine, a HIV other antiretroviral therapy compound and combinations thereof, or a pharmaceutically acceptable salt of any of the aforementioned.
  • NRTI non-nucleoside reverse transcriptase inhibitor
  • NRTI nucleoside reverse transcriptase inhibitor
  • PI protease inhibitor
  • INSTI integrase strand transfer inhibitor
  • HIV vaccine a HIV other antiretroviral therapy compound and combinations thereof, or a pharmaceutically acceptable salt of any of the aforementioned.
  • the subject suffering from the HIV infection is not using an agent selected from a non-nucleoside reverse transcriptase inhibitor (NNRTI), a nucleoside reverse transcriptase inhibitor (NRTI), a protease inhibitor (PI), a fusion/entry inhibitor, an integrase strand transfer inhibitor (INSTI), a HIV vaccine, a HIV other antiretroviral therapy compound and combinations thereof, or a pharmaceutically acceptable salt of any of the aforementioned.
  • NRTI non-nucleoside reverse transcriptase inhibitor
  • NRTI nucleoside reverse transcriptase inhibitor
  • PI protease inhibitor
  • INSTI integrase strand transfer inhibitor
  • HIV vaccine a HIV other antiretroviral therapy compound and combinations thereof, or a pharmaceutically acceptable salt of any of the aforementioned.
  • NNRTIs include, but are not limited to, delavirdine (Rescriptor®), efavirenz (Sustiva®), etravirine (Intelence®), nevirapine (Viramune®), rilpivirine (Edurant®), doravirine, and pharmaceutically acceptable salts of any of the foregoing, and/or a combination thereof.
  • Suitable NRTIs include, but are not limited to, abacavir (Ziagen®), adefovir (Hepsera), amdoxovir, apricitabine, censavudine, didanosine (Videx®), elvucitabine, emtricitabine (Emtriva®), entecavir (Baraclude®), lamivudine (Epivir®), racivir, stampidine, stavudine (Zerit®), tenofovir disoproxil (including Viread®), tenofovir alafenamide, zalcitabine (Hivid®), zidovudine (Retrovir®), and pharmaceutically acceptable salts of any of the foregoing, and/or a combination thereof.
  • abacavir Ziagen®
  • Hepsera adefovir
  • Amdoxovir apricitabine
  • censavudine didanosine
  • vaccines include, but are not limited to Heplislav®, ABX-203, INO-1800, and pharmaceutically acceptable salts of any of the foregoing, and/or combinations thereof.
  • suitable protease inhibitors include, but are not limited to, atazanavir (Reyataz®), darunavir (Prezista®), fosamprenavir (Lexiva®), indinavir (Crixivan®), lopinavir/ritonavir (Kaletra), nelfinavir (Viracept®), ritonavir (Norvir®) and saquinavir (Invirase®).
  • fusion/entry inhibitors include, but are not limited to, enfuvirtide (Fuzeon®), maraviroc (Selzentry®), vicriviroc, apliviroc, ibalizumab, fostemsavir and PRO-140.
  • suitable INSTIs include, but are not limited to, raltegravir (Isentress®), dolutegravir (Tivicay®) and elvitegravir (Viteka®).
  • HIV-1 is more virulent and more infective strain, and has a global prevalence. HIV-2 is considered to be less virulent and is geographically confined.
  • an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof can be effective to treat HIV-1.
  • an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof can be effective to treat HIV-2.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • Suitable indicators include, but are not limited to, a reduction in viral load, a reduction in plasma viral load, a reduction in viral replication, a reduction in time to seroconversion (virus undetectable in patient serum), an increase CD4+T lymphocyte counts, a reduction in the population of HIV infected cells, a reduction of morbidity or mortality in clinical outcomes and/or a reduction in the rate of opportunistic infections.
  • successful therapy with an effective amount of a compound or a pharmaceutical composition described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and an effective amount of a HIV latency reversing agent, or a pharmaceutically acceptable salt thereof can reduce the incidence of opportunistic infections in HIV infected subjects.
  • an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof is an amount that is effective to reduce the population of CD4+T lymphocyte cells harboring HIV to undetectable levels.
  • an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof is an amount that is effective to increase CD4+ T lymphocyte counts from less than about 200 cells/mL to greater than about 1,200 cells/mL. In some embodiments, an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, is an amount that is effective to increase CD4+T lymphocyte counts from less than about 200 cells/mL to greater than about 500 cells/mL.
  • infectious agents can develop resistance to one or more therapeutic agents.
  • resistance refers to a viral strain displaying a delayed, lessened and/or null response to a therapeutic agent(s).
  • the virus sometimes mutates or produces variations that are resistant or partially resistant to certain drugs.
  • the viral load of a subject infected with a resistant virus may be reduced to a lesser degree compared to the amount in viral load reduction exhibited by a subject infected with a non-resistant strain.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can be provided to a subject infected with a HIV strain that is resistant to one or more different anti-HIV agents (for example, an agent used in a conventional standard of care).
  • anti-HIV agents include, but are not limited to, non-nucleoside reverse transcriptase inhibitors (NNRTIs), nucleoside reverse transcriptase inhibitors (NRTIs), protease inhibitors (PIs), fusion/entry inhibitors, integrase strand transfer inhibitors (INSTIs), HIV vaccines, and combinations thereof, or pharmaceutically acceptable salts of any of the aforementioned.
  • Bcl-2 inhibitors can cause one or more undesirable side effects in the subject being treated.
  • undesirable side effects include, but are not limited to, thrombocytopenia, neutropenia, anemia, diarrhea, nausea and upper respiratory tract infection.
  • a compound described herein for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof can result in a severity of a side effect (such as one of those described herein) that is 25% less than compared to the severity of the same side effect experienced by a subject receiving a known Bcl-2 inhibitors (such as venetoclax, navitoclax, obatoclax, ABT-737, S55746, AT-101, APG-1252 and APG-2575).
  • a side effect such as one of those described herein
  • a side effect such as one of those described herein
  • a known Bcl-2 inhibitors such as venetoclax, navitoclax, obatoclax, ABT-737, S55746, AT-101, APG-1252 and APG-2575.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof results in a number of side effects that is 25% less than compared to the number of side effects experienced by a subject receiving a known Bcl-2 inhibitors (for example, venetoclax, navitoclax, obatoclax, ABT-737, S55746, AT-101, APG-1252 and APG-2575).
  • a known Bcl-2 inhibitors for example, venetoclax, navitoclax, obatoclax, ABT-737, S55746, AT-101, APG-1252 and APG-2575.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof results in a severity of a side effect (such as one of those described herein) that is less in the range of about 10% to about 30% compared to the severity of the same side effect experienced by a subject receiving a known Bcl-2 inhibitors (for example, venetoclax, navitoclax, obatoclax, ABT-737, S55746, AT-101, APG-1252 and APG-2575).
  • a side effect such as one of those described herein
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof results in a number of side effects that is in the range of about 10% to about 30% less than compared to the number of side effects experienced by a subject receiving a known Bcl-2 inhibitors (for example, venetoclax, navitoclax, obatoclax, ABT-737, S55746, APG-1252 and APG-2575).
  • a known Bcl-2 inhibitors for example, venetoclax, navitoclax, obatoclax, ABT-737, S55746, APG-1252 and APG-2575.
  • the one or more compounds of Formula (I), or a pharmaceutically acceptable salt thereof, that can be used to treat, ameliorate and/or inhibit the replication of a cancer, malignant growth, or tumor wherein inhibiting the activity of Bcl-2 is beneficial is provided in any of the embodiments described above under the heading titled “Compounds.”
  • the methods and uses described above in the Uses and Methods of Treatment section of this disclosure are carried out in the described manner (generally involving cancer, malignant growth, and/or tumor) using a compound of Formula (I) in which R 3 is hydrogen or halogen, or a pharmaceutically acceptable salt thereof.
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving cancer, malignant growth, and/or tumor) using a compound of Formula (I) in which R 3 is X—R 3A ,
  • the methods and uses described above in the Uses and Methods of Treatment section of this disclosure are carried out in the described manner (generally involving cancer, malignant growth, and/or tumor) using a compound of Formula (I) in which R 3 is X—R 3A ,
  • X 2 are —NH—.
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving cancer, malignant growth, and/or tumor) using a compound of Formula (I) in which R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine, with the proviso that R 1 is not —CH 2 F, —CHF 2 or —CF 3 ; R 3 is X—R 3A ,
  • X 1 and X 2 are —NH—.
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving cancer, malignant growth, and/or tumor) using a compound of Formula (I) in which R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine;
  • R 3 is X—R 3A ,
  • X 1 and X 2 are —O—.
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving cancer, malignant growth, and/or tumor) using a compound of Formula (I) in which R 1 is —CH 2 F, —CHF 2 or —CF 3 ; R 3 is X—R 3A ,
  • X 1 and X 2 are —NH—.
  • the one or more compounds of Formula (I), or a pharmaceutically acceptable salt thereof, that can be used to treat and/or ameliorate a HIV infection and/or reducing the reoccurrence of a HIV infection and/or reducing the population of HIV infected cells, wherein inhibiting the activity of Bcl-2 is beneficial is provided in any of the embodiments described above under the heading titled “Compounds.”
  • the methods and uses described above in the Uses and Methods of Treatment section of this disclosure are carried out in the described manner (generally involving HIV) using a compound of Formula (I) in which R 3 is hydrogen or halogen, or a pharmaceutically acceptable salt thereof.
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving HIV) using a compound of Formula (I) in which R 3 is X—R 3A ,
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving HIV) using a compound of Formula (I) in which R 1 is selected from the group consisting of hydrogen, halogen, a substituted or unsubstituted C 1 -C 6 alkyl, a substituted or unsubstituted C 1 -C 6 haloalkyl, a substituted or unsubstituted C 3 -C 6 cycloalkyl, a substituted or unsubstituted C 1 -C 6 alkoxy, an unsubstituted mono-C 1 -C 6 alkylamine and an unsubstituted di-C 1 -C 6 alkylamine, with the proviso that R 1 is not —CH 2 F, —CHF 2 or —CF 3 ; R 3 is X—R 3A ,
  • X 1 and X 2 are —NH—.
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving HIV) using a compound of Formula (I) in which R 3 is X—R 3A ,
  • the methods and uses described above in the Uses and Methods of Treatment section are carried out in the described manner (generally involving HIV) using a compound of Formula (I) in which R 1 is —CH 2 F, —CHF 2 or —CF 3 ; R 3 is X—R 3A ,
  • X 2 are —NH—.
  • a “subject” refers to an animal that is the object of treatment, observation or experiment.
  • Animal includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals.
  • “Mammal” includes, without limitation, mice, rats, rabbits, guinea pigs, dogs, cats, sheep, goats, cows, horses, primates, such as monkeys, chimpanzees, and apes, and, in particular, humans.
  • the subject can be human.
  • the subject can be a child and/or an infant, for example, a child or infant with a fever.
  • the subject can be an adult.
  • treatment does not necessarily mean total cure or abolition of the disease or condition. Any alleviation of any undesired signs or symptoms of the disease or condition, to any extent can be considered treatment and/or therapy.
  • treatment may include acts that may worsen the subject's overall feeling of well-being or appearance.
  • a therapeutically effective amount of compound, salt or composition can be the amount needed to prevent, alleviate or ameliorate symptoms of the disease or condition, or prolong the survival of the subject being treated. This response may occur in a tissue, system, animal or human and includes alleviation of the signs or symptoms of the disease or condition being treated. Determination of an effective amount is well within the capability of those skilled in the art, in view of the disclosure provided herein.
  • the therapeutically effective amount of the compounds disclosed herein required as a dose will depend on the route of administration, the type of animal, including human, being treated and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.
  • an effective amount of a compound is the amount that results in: (a) the reduction, alleviation or disappearance of one or more symptoms caused by the cancer, (b) the reduction of tumor size, (c) the elimination of the tumor, and/or (d) long-term disease stabilization (growth arrest) of the tumor.
  • a therapeutically effective amount is that amount that alleviates or eliminates cough, shortness of breath and/or pain.
  • an effective amount, or a therapeutically effective amount of a Bcl-2 inhibitor is the amount which results in the reduction in Bcl-2 activity and/or an increase in apoptosis.
  • the reduction in Bcl-2 activity is known to those skilled in the art and can be determined by the analysis of Bcl-2 binding and relatives levels of cells undergoing apoptosis.
  • the amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, required for use in treatment will vary not only with the particular compound or salt selected but also with the route of administration, the nature and/or symptoms of the disease or condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. In cases of administration of a pharmaceutically acceptable salt, dosages may be calculated as the free base. As will be understood by those of skill in the art, in certain situations it may be necessary to administer the compounds disclosed herein in amounts that exceed, or even far exceed, the dosage ranges described herein in order to effectively and aggressively treat particularly aggressive diseases or conditions.
  • a suitable dose will often be in the range of from about 0.05 mg/kg to about 10 mg/kg.
  • a suitable dose may be in the range from about 0.10 mg/kg to about 7.5 mg/kg of body weight per day, such as about 0.15 mg/kg to about 5.0 mg/kg of body weight of the recipient per day, about 0.2 mg/kg to 4.0 mg/kg of body weight of the recipient per day, or any amount in between.
  • the compound may be administered in unit dosage form; for example, containing 1 to 500 mg, 10 to 100 mg, 5 to 50 mg or any amount in between, of active ingredient per unit dosage form.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations.
  • the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight, the severity of the affliction, the mammalian species treated, the particular compounds employed and the specific use for which these compounds are employed.
  • the determination of effective dosage levels can be accomplished by one skilled in the art using routine methods, for example, human clinical trials, in vivo studies and in vitro studies.
  • useful dosages of a compound of Formula (I), or pharmaceutically acceptable salts thereof can be determined by comparing their in vitro activity and in vivo activity in animal models. Such comparison can be done by comparison against an established drug, such as cisplatin and/or gemcitabine)
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vivo and/or in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations. Dosage intervals can also be determined using MEC value.
  • Compositions should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
  • the attending physician would know how to and when to terminate, interrupt or adjust administration due to toxicity or organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the disease or condition to be treated and to the route of administration. The severity of the disease or condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the age, body weight and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
  • the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties may be established by determining in vitro toxicity towards a cell line, such as a mammalian, and preferably human, cell line. The results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • a cell line such as a mammalian, and preferably human, cell line.
  • the results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans.
  • the toxicity of particular compounds in an animal model such as mice, rats, rabbits, dogs or monkeys, may be determined using known methods.
  • the efficacy of a particular compound may be established using several recognized methods, such as in vitro methods, animal models, or human clinical trials. When selecting a model to determine efficacy, the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, route of administration and/or regime.
  • Step 1 A solution of 2-oxa-8-azaspiro[4.5] decane hydrochloride (500 mg, 2.81 mmol) in CH 3 CN (20 mL) was treated with tert-butyl-2-bromoethylcarbamate (700 mg, 3.12 mmol) and K 2 CO 3 (1.55 g, 11.24 mmol) and heated to 80° C. for 16 h. The reaction was concentrated, diluted with water (20 mL), and extracted with EtOAc (3 ⁇ 20 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The residue was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 2 To a stirred solution of Intermediate 4-1 (500 mg, 1.76 mmol) in DCM (20 mL) was added HCl (4 M in dioxane, 10 mL) at 0° C. The reaction was warmed to rt, stirred for 2 h, concentrated and triturated with Et 2 O to afford 2-(2-oxa-8-azaspiro[4.5]decan-8-yl)ethanamine dihydrochloride (Intermediate 4-2) (300 mg, 66%) as an off white solid which was used for the next step without further purification.
  • Step 3 A solution of Intermediate 4-2 (300 mg, 1.17 mmol) in CH 3 CN (15 mL) was treated with 4-chloro-3-nitrobenzenesulfonamide (276 mg, 1.17 mmol) followed by DIPEA (0.82 mL, 4.68 mmol) and then heated to 80° C. After 16 h, the reaction was cooled to rt and concentrated. The crude product was purified by column chromatography (SiO 2 , MeOH (0.1% triethylamine)/DCM) to afford Intermediate 4 (300 mg, 66%) as a yellow solid. LC/MS (ESI) m/z 385.3 [M+H]+.
  • Step 1 tert-butyl 2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)ethylcarbamate (Intermediate 5-1) was prepared following the procedure described in Step 1 for Intermediate 4 using 7-oxa-2-azaspiro[3.5] nonane hemioxalic acid in place of 2-oxa-8-azaspiro[4.5] decane hydrochloride 1 H NMR (300 MHz, DMSO-d 6 ) ⁇ 6.94 (br s, 1H), 3.74 (br s, 4H), 3.51-3.42 (m, 4H), 3.10 (br s, 4H), 1.76 (br s, 4H), 1.39 (s, 9H).
  • Step 2 2-(7-oxa-2-azaspiro[3.5]nonan-2-yl)ethanamine dihydrochloride (Intermediate 5-2) was prepared following the procedure described in Step 2 for Intermediate 4 using Intermediate 5-1 in place of Intermediate 4-1.
  • 1 H NMR 300 MHz, DMSO-d 6 ) ⁇ 11.42 (br s, 1H), 8.3 (br s, 3H), 4.05-3.99 (m, 2H), 3.92-3.86 (m, 2H), 3.57-3.54 (m, 4H), 3.49-3.40 (m, 4H), 3.10-3.05 (m, 2H), 1.88 (br s, 2H), 1.72 (br s, 2H).
  • Step 3 A solution of Intermediate 5-2 (250 mg, 1.03 mmol) in CH 3 CN (13 mL) was treated with 4-fluoro-3-nitrobenzenesulfonamide (226.8 mg, 1.03 mmol) followed by triethylamine (0.58 mL, 4.12 mmol) at rt. After 16 h, the reaction was concentrated to afford the crude product, which was purified by column chromatography (SiO 2 , MeOH (containing 7N NH 3 )/DCM) to obtain Intermediate 5 (200 mg, 52%) as a yellow solid. LC/MS (ESI) m/z 371.3 [M+H]+.
  • Step 1 To a stirred solution of 1-(3-hydroxy-2-(tetrahydro-2H-pyran-2-yloxy)propyl)cyclopropanol (prepared according to CN106565706) and triphenyl phosphine (9.10 g, 34.7 mmol) in THF (50 mL), was added diethyl azodicarboxylate (DEAD) (5.44 mL, 34.7 mmol) dropwise at rt. After 16 h, the reaction mixture was quenched with H 2 O (50 mL) and extracted with EtOAc (3 ⁇ 50 mL). The combined organic layers were washed with water (50 mL), dried over Na 2 SO 4 and concentrated.
  • DEAD diethyl azodicarboxylate
  • Step 2 To a stirred solution of Intermediate 7-1 (3.2 g, 16.1 mmol) in MeOH (32 mL) was added pyridinium p-toluenesulfonate (811 mg, 3.23 mmol) and stirred at 40° C. for 5 h. The reaction mixture was concentrated, and the residue was purified by column chromatography (SiO 2 , EtOAc/pet. ether) to obtain 4-oxaspiro[2.4]heptan-6-ol (Intermediate 7-2) (1.0 g, 54% yield) as colorless oil. GC/MS m/z 114.1 [M]+.
  • Step 3 To a stirred solution of Intermediate 7-2 was added sodium hydride (63% dispersion in oil, 1.05 g, 26.3 mmol) at 0° C. After 30 min, a solution of 4-fluoro-3-nitrobenzenesulfonamide (1.92 g, 8.76 mmol) in THF (5 mL) was added dropwise at 0° C. The reaction was warmed to rt and stirred for 6 h. The reaction was cooled to 0° C. and quenched with sat. aq. NH 4 Cl and extracted with EtOAc (3 ⁇ 50 mL). The combined organic layers were dried over Na 2 SO 4 and concentrated. The residue was triturated with Et 2 O and n-pentane to afford Intermediate 7 (700 mg, 25% yield) as a white solid. LC/MS (ESI) m/z 313.0 [M ⁇ H] ⁇ .
  • Step 1 To a stirred solution of 3-(methoxycarbonyl)bicyclo[1.1.1]pentane-1-carboxylic acid (10 g, 58.8 mmol), N,O-dimethylhydroxylamine hydrochloride (6.88 g, 42.4 mmol) and triethylamine (12.3 mL, 176.4 mmol) in DCM (200 mL) at 0° C. was added T 3 P (50% solution in EtOAc, 18.8 g, 58.8 mmol). The resulting reaction mixture warmed to rt and stirred for 16 h. The reaction mixture was quenched with water (250 mL) and extracted with DCM (3 ⁇ 250 mL).
  • Step 2 To a stirred solution of Intermediate 13-1 (5 g, 23.5 mmol) in THF (100 mL) at ⁇ 78° C. was added MeMgBr (3M in Et 2 O, 31.3 mL, 93.8 mmol). After stirring for 2 h at ⁇ 78° C., the reaction was quenched with sat. aq. NH 4 Cl (100 mL) and extracted with EtOAc (3 ⁇ 100 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 3 A solution of the Intermediate 13-2 (2.3 g, 13.6 mmol) in DCM (50 mL) at ⁇ 78° C. was treated dropwise with DAST (6.62 g, 41.0 mmol). After the addition, the temperature was raised to rt. After 16 h, the reaction mixture was cooled to ⁇ 78° C. and carefully quenched with sat. aq. NaHCO 3 (100 mL). The mixture was extracted with DCM (3 ⁇ 100 mL) and the combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by chromatography chromatography (SiO 2 , EtOAc/pet.
  • Step 4 To a stirred solution of Intermediate 13-3 (1.8 g, 9.46 mmol) and N,O-dimethylhydroxylamine hydrochloride (0.923 g, 9.46 mmol) in anhydrous THF (40 mL) at ⁇ 78° C. was added i-PrMgCl (2M in THF, 18.9 mL, 37.8 mmol). The reaction mixture was warmed ⁇ 50° C. and stirred for 2 h. The reaction mixture was quenched with sat. aq. NH 4 Cl (50 mL) and extracted with EtOAc (3 ⁇ 75 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated.
  • Step 1 To a stirred solution of tert-butyl 4-(aminomethyl)-4-fluoropiperidine-1-carboxylate (2.00 g, 8.61 mmol) in THF (30 mL), was added 4-fluoro-3-nitrobenzenesulfonamide (2.08 g, 9.47 mmol) followed by triethylamine (4.8 mL, 34.45 mmol). The resulting reaction mixture was stirred at rt for 16 h. The reaction was then concentrated, and the resulting residue was diluted with 10% MeOH-DCM (50 mL) and washed with ice-cold water (5 ⁇ 50 mL). The organic layer was dried over Na 2 SO 4 , filtered and concentrated.
  • Step 2 To a stirred solution of Intermediate 15-1 (1.6 g, 3.70 mmol) in 1,4-dioxane (10 mL) at 0° C. was added HCl (4M HCl in 1,4-dioxane, 20 mL). The reaction was warmed to rt and stirred for 6 h. The reaction was concentrated and triturated with Et 2 O to afford 4-(((4-fluoropiperidin-4-yl)methyl)amino)-3-nitrobenzenesulfonamide hydrochloride (Intermediate 15-2) (1.3 g, 96%) as a yellow solid. LC/MS (ESI) m/z 333.1 [C 12 H 17 FN 4 O 4 S+H] + .
  • Step 3 To a stirred solution of Intermediate 15-2 (430 mg, 1.35 mmol) in MeOH (15 mL) was added paraformaldehyde (81 mg, 2.71 mmol) at 0° C. After 15 min, NaCNBH 3 (128 mg, 2.03 mmol) was added and the reaction was warmed to rt. After 18h, the reaction was quenched sat. aq. NaHCO 3 (15 mL) and the reaction was extracted with DCM (3 ⁇ 100 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was triturated with Et 2 O followed by 1:1 EtOAc/Hexane to afford Intermediate 15 (340 mg, 25% yield) as a yellow solid. LC/MS (ESI) m/z 347.1 [M+H] + .
  • the absolute stereochemistry was arbitrarily assigned for Intermediate 17A.
  • LC/MS (ESI) m/z 331.2 [M+H] + .
  • the absolute stereochemistry was arbitrarily assigned for Intermediate 17B.
  • Step 1 A solution of NaClO 2 (11.08 g, 122.5 mmol) in water (100 mL) was added drop wise to a stirring mixture of 2-bromo-4,4-dimethylcyclohex-1-ene-1-carbaldehyde (19 g, 87.5 mmol), CH 3 CN (100 mL), NaH 2 PO 4 (2.72 g, 22.75 mmol), water (40 mL) and 30% H 2 O 2 (aq.) (15 mL) at 10° C. Upon completion, the reaction, was poured into sat. aq. Na 2 CO 3 (200 mL) and washed with Et 2 O (200 mL).
  • Step 2 To a stirred solution of Intermediate 20-1 (10 g, 42.9 mmol) in DMF (100 mL) was added K 2 CO 3 (17.79 g, 128.7 mmol) followed by benzyl bromide (14.67 g, 85.8 mmol) at 0° C. and the reaction was warmed to rt. After 16 h, water (200 mL) was added and the reaction was extracted with EtOAc (3 ⁇ 200 mL). The combined organic layers were washed with water (3 ⁇ 200 mL), dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 1 A stirring solution of methyl 3-formylbicyclo[0.1.1]pentane-1-carboxylate (7.5 g, 48.7 mmol) in DCM (100 mL) was cooled to ⁇ 78° C., and treated with DAST (19.3 mL, 146.1 mmol) drop wise and warmed to rt. After 6 h, the reaction mixture was cooled to ⁇ 78° C. and quenched with sat. aq. NaHCO 3 (100 mL) and extracted with DCM (3 ⁇ 100 mL).
  • Step 2 To a stirred solution of Intermediate 21-1 (7 g, 39.74 mmol) in anhydrous THF (70 mL) was added N,O-dimethylhydroxylamine hydrochloride (3.89 g, 39.74 mmol) at ⁇ 78° C., followed by i-PrMgCl (2M in THF, 79.5 mL, 159 mmol). The reaction was warmed to ⁇ 50° C. and stirred for 2 h. The reaction mixture was then quenched with sat. aq. NH 4 Cl solution (100 mL) and extracted with EtOAc (3 ⁇ 100 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated.
  • Step 1 A solution of 1-iodo-3-methylbicyclo[1.1.1]pentane (30 g, 144.20 mmol) in THF (225 mL) was cooled to ⁇ 78° C. and sec-butyllithium (1.4M in cyclohexane, 154.50 mL, 216.30 mmol) was added drop wise over 1 h. The resulting pale yellow suspension was stirred at ⁇ 78° C. for 10 min and then warmed to 0° C. and stirred for 80 min. The reaction mixture was then cooled to ⁇ 78° C., and a solution of Intermediate 19 (24.67 g, 108.15 mmol) in THF (75 mL) was added drop wise over 20 min.
  • Step 2 A solution of Intermediate 22-1 (62 g, 199.69 mmol) in 1,4-dioxane (1.24 L), was treated with 2N HCl(aq.) (299.5 mL, 599.2 mmol) at rt and then warmed to 70° C. After 16 h, the reaction was cooled to rt, poured into water (1.24 L) and extracted with Et 2 O (2 ⁇ 750 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet. ether) to provide Intermediate 22 (23 g, 36% yield over 2 steps) as a yellow oil.
  • Step 1 To a stirred solution of [0.1.11]propellane (0.19M in Et 2 O/pentane), 128.6 mmol) at ⁇ 78° C. was added EtI (18.7 g, 257.38 mmol). The reaction was warmed to rt and stirred for 3 days in the dark. The reaction was then concentrated at 0° C. to afford 1-ethyl-3-iodobicyclo[1.1.1]pentane (Intermediate 23-1) (21.2 g, 74% yield) as yellow oil.
  • Step 2 To a stirred solution of Intermediate 23-1 (10.90 g, 49.1 mmol) in Et 2 O (75 mL) at ⁇ 78° C. was added sec-BuLi (1.4 M in cyclohexane, 50 mL, 70.0 mmol). After 10 min, the reaction was warmed to rt and stirred for 1 h. The reaction mixture was then cooled to ⁇ 78° C. and treated with a solution of 2-(diethoxymethyl)-5,5-dimethylcyclohexan-1-one (8 g, 35.0 mmol) in Et 2 O (25 mL). After 1 h, the reaction was warmed to 0° C. and stirred for 2 h. The reaction was quenched with sat.
  • Step 3 A solution of Intermediate 23-2 (8.5 g, crude) in acetone (80 mL), was treated with 2N HCl(aq.) (20 mL) at rt and then warmed to 75° C. After 24 h, the reaction was concentrated and then diluted with water (50 mL) and extracted with Et 2 O (3 ⁇ 250 mL). The combined organic layers were washed with sat. aq. NaHCO 3 , dried over Na 2 SO 4 and concentrated. The crude product was purified by column chromatography (SiO 2 , Et 2 O/pet. ether) to provide Intermediate 23 (3.9 g, 48% yield over 2 steps) as a brown oil.
  • Step 1 Preparation of CF 2 HI (based on a procedure from Cao, P. et. al. J. Chem. Soc., Chem. Commun. 1994, 737-738): performed in two parallel batches: A mixture of KI (94 g, 568 mol), MeCN (228 ml) and water (18 mL) was heated to 45° C. and treated with, 2,2-difluoro-2-(fluorosulfonyl)acetic acid (50 g, 284 mmol) in MeCN (50 mL) dropwise over 4 h. The reaction mixture was then cooled to 0° C., and diluted with pentane (150 mL) and water (125 mL).
  • Step 2 To a stirred solution of [0.1.1]propellane (0.53 M in Et 2 O, 52 mL, 27.56 mmol) at ⁇ 40° C. was added Intermediate 24-1 (0.15 M in pentane, 200 mL, 30 mmol). The reaction mixture was warmed to rt, protected from light, and stirred for 2 days. The reaction was then concentrated at 0-10° C. to obtain 1-(difluoromethyl)-3-iodobicyclo[1.1.1]pentane (Intermediate 24-2) (5 g, 20.5 mmol, 74% yield) as a white solid.
  • Step 3 A solution of Intermediate 24-2(30 g, 122.94 mmol) in THF (225 mL) was cooled to ⁇ 78° C. and sec-butyllithium (1.4M in cyclohexane, 219 mL, 306.7 mmol) was added drop-wise for 1 h. The resulting pale yellow suspension was stirred at ⁇ 78° C. for 10 min and temperature was raised to 0° C. and stirred for 80 min. The reaction mixture was then cooled to ⁇ 78° C., and a solution of Intermediate 19 (21 g, 92.20 mmol) in THF (75 mL) was added drop wise to the reaction over 20 min. After 10 min, the reaction was warmed to 0° C.
  • Step 4 Intermediate 24 was prepared following the procedure described in Step 2 for Intermediate 22 using Intermediate 24-3 in place of Intermediate 22-1 (38% over 2 steps).
  • Step 1 To a stirred solution of 1-iodo-3-(trifluoromethyl)bicyclo[1.1.1]pentane (5.00 g, 19.1 mmol) in Et 2 O (100 mL) at ⁇ 78° C. was added sec-BuLi (1.4 M in cyclohexane, 13.63 mL, 19.08 mmol. After 10 minutes at ⁇ 78° C., the reaction was warmed to 0° C. and stirred for 1 h. The reaction mixture was then cooled to ⁇ 78° C. and then a solution of Intermediate 19 (3.63 g, 15.90 mmol) in Et 2 O (50 mL) was added. After 1 h, the reaction was warmed to 0° C.
  • Step 2 Intermediate 25 was prepared following the procedure described in Step 3 for Intermediate 23 using Intermediate 25-1 in place of Intermediate 23-2.
  • Step 1 A solution of 5-iodo-4,4-dimethylpent-1-ene (9.85 g, 44.0 mmol) in pentane (100 mL) was treated with t-BuLi (64.6 mL, 1.7 M in n-pentane, 109.9 mmol) at ⁇ 78° C. under inert atmosphere. After 1 h, a solution of Intermediate 1 (5 g, 26.4 mmol) in THF (20 mL) was added and the mixture was stirred at ⁇ 78° C. for 1 h. The reaction was then warmed to ⁇ 30° C. over 30 min. and stirred for 1 h. The reaction was quenched with sat. aq.
  • Step 2 A solution of Intermediate 26-1 (3.1 g, 13.7 mmol) and acrylonitrile (2.18 g, 41.0 mmol) in degassed DCM (120 mL) was treated dropwise over 2 h with a solution of Hoveyda-Grubbs CatalystTM 2nd Generation (343 mg, 0.55 mmol) in DCM (5 mL) at 45° C. The reaction was stirred at 45° C. for 48 h, cooled to rt, concentrated and absorbed onto Celite. The residue was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 3 A solution of Intermediate 26-2 (700 mg, 2.78 mmol) in MeOH (20 mL) was treated with Pd/C (10 wt %, 170 mg) and stirred under an atmosphere of H 2 (1 atm) for 2 h. The reaction was purged with N 2 and the reaction mixture was filtered over Celite and concentrated to provide 7-(3-chlorobicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxoheptanenitrile (Intermediate 26-3) (550 mg, 77%) as a clear colorless oil.
  • Step 4 A solution of Intermediate 26-3 (1.1 g, 4.34 mmol, 1 eq) in THF (20 mL) was treated with 4 ⁇ molecular sieves (100 mg) and 15-Crown-5 (956 mg, 4.34 mmol) and was placed in a preheated 70° C. oil bath. After 2 min, the reaction was treated with t-BuONa (2.09 g, 21.7 mmol) in a single portion. After 5 h, the reaction was cooled to rt and poured into a stirring solution of sat. aq. NH 4 Cl. The aqueous phase was washed with DCM (3 ⁇ 25 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated.
  • Step 5 To a stirred solution of Intermediate 26-4 (400 mg, 1.70 mmol) in anhydrous DCM (20 mL) at ⁇ 78° C. was added DIBAL-H (2.55 mL, 1M in toluene, 2.55 mmol). The reaction was warmed to rt. After 4 h, the reaction was cooled to 0° C., quenched with 2M HCl(aq.) (40 mL) and warmed to rt.
  • DIBAL-H 2.55 mL, 1M in toluene, 2.55 mmol
  • Step 6 To a stirred solution of Intermediate 26-5 (300 mg, 1.26 mmol) in DCM (10 mL) was added Intermediate 2 (544 mg, 1.38 mmol) and NaBH(OAc) 3 (347 mg, 1.64 mmol) at rt. After 16 h, additional NaBH(OAc) 3 (347 mg, 1.64 mmol) was added. After 48 h, the reaction was quenched with MeOH (0.2 mL) at 0° C., warmed to rt and concentrated. The residue was diluted with DCM and washed with sat. aq. NaHCO 3 .
  • Step 7 To a solution of Intermediate 26-6 (125 mg, 0.20 mmol) in DCM (2 mL) at 0° C. was added TFA (139 mg, 1.22 mmol). The mixture was warmed to rt and stirred for 3 h and concentrated to provide the TFA salt of 2-(1H-pyrrolo[2,3-b]pyridin-5-yl-oxy)-4-(4-((2-(3-chlorobicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-yl)methyl)piperazin-1-yl)benzoic acid (140 mg, quantitative) as a white solid LC/MS (ESI) m/z 561.3 [C 32 H 37 ClN 4 O 3 +H] + .
  • Step 1 1-(3-fluorobicyclo[1.1.1]pentan-1-yl)-3,3-dimethylhex-5-en-1-one (Intermediate 27-1) was prepared following the procedure described in Step 1 for Intermediate 26 using Intermediate 11 in place of Intermediate 1.
  • Step 2 E/Z-7-(3-fluorobicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxohept-2-enenitrile (Intermediate 27-2) was prepared following the procedure described in Step 2 for Intermediate 26 using Intermediate 27-1 in place of Intermediate 26-1.
  • LC/MS (ESI) m/z 236.3 [M+H] + .
  • Step 3 7-(3-fluorobicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxoheptanenitrile (Intermediate 27-3) was prepared following the procedure described in Step 3 for Intermediate 26 using Intermediate 27-2 in place of Intermediate 26-2.
  • Step 4 2-(3-fluorobicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enecarbonitrile (Intermediate 27-4) was prepared following the procedure described in Step 4 for Intermediate 26 using Intermediate 27-3 in place of Intermediate 26-3.
  • LC/MS (ESI) m/z 220.4 [M+H] + .
  • Step 5 2-(3-fluorobicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enecarbaldehyde (Intermediate 27-5) was prepared following the procedure described in Step 5 for Intermediate 26 using Intermediate 27-4 in place of Intermediate 26-4.
  • 1 H NMR 300 MHz, CDCl 3 ) ⁇ 10.19 (s, 1H), 2.37-2.34 (m, 6H), 2.30-2.25 (m, 2H), 1.93 (br s, 2H), 1.40-1.35 (m, 2H), 0.91 (s, 6H).
  • Step 6 To a stirred solution of Intermediate 27-5 (100 mg, 0.45 mmol) in EtOH (4 mL) was added Intermediate 2 (195 mg, 0.49 mmol) and AcOH (cat.) at rt and stirred for 15 min. The resulting reaction mixture was cooled to 0° C. and NaCNBH 3 (42 mg, 0.675 mmol) was added and the reaction was warmed to rt. After 16 h, the reaction was concentrated and the residue was diluted with sat. aq. NaHCO 3 (10 ml) and extracted with DCM (3 ⁇ 10 ml). The combined organic layers were dried over Na 2 SO 4 and concentrated. The crude compound was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 7 2-(1H-pyrrolo[2,3-b]pyridin-5-yl-oxy)-4-(4-((2-(3-fluorobicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-yl)methyl)piperazin-1-yl)benzoic acid as the TFA salt was prepared following the procedure described in Step 7 for Intermediate 26 by reacting Intermediate 27-6 in place of Intermediate 26-6.
  • LC/MS (ESI) m/z 545.4 [C 32 H 37 FN 4 O 3 +H] + .
  • Step 1 3,3-dimethyl-1-(3-methylbicyclo[1.1.1]pentan-1-yl)hex-5-en-1-one (Intermediate 28-1) was prepared following the procedure described in Step 1 for Intermediate 26 using Intermediate 10 in place of Intermediate 1.
  • Step 2 E/Z-5,5-dimethyl-7-(3-methylbicyclo[1.1.1]pentan-1-yl)-7-oxohept-2-enenitrile (Intermediate 28-2) was prepared following the procedure described in Step 2 for Intermediate 26 using Intermediate 28-1 in place of Intermediate 26-1.
  • LC/MS (ESI) m/z 232.3 [M+H] + .
  • Step 3 5,5-dimethyl-7-(3-methylbicyclo[1.1.1]pentan-1-yl)-7-oxoheptanenitrile (Intermediate 28-3) was prepared following the procedure described in Step 3 for Intermediate 26 using Intermediate 28-2 in place of Intermediate 26-2.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 2.33-2.29 (m, 4H), 1.86 (s, 6H), 1.64-1.56 (m, 2H), 1.50-1.45 (m, 2H), 1.18 (s, 3H), 0.98 (s, 6H).
  • Step 4 4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-enecarbonitrile (Intermediate 28-4) was prepared following the procedure described in Step 4 for Intermediate 26 using Intermediate 28-3 in place of Intermediate 26-3.
  • LC/MS (ESI) m/z 216.4 [M+H] + .
  • Step 5 Intermediate 22 was prepared following the procedure described in Step 5 for Intermediate 26 using Intermediate 28-4 in place of Intermediate 26-4. LC/MS (ESI) m/z 219.3 [M+H] + .
  • Step 6 To a stirred solution of Intermediate 22 (70 mg, 0.32 mmol) in EtOH (4 mL) was added Intermediate 2 (190 mg, 0.48 mmol) and AcOH (cat.) at rt. After 15 min, the mixture was cooled to 0° C., NaCNBH 3 (31 mg, 0.48 mmol) was added and the reaction was warmed to rt. After 16 h, the reaction was concentrated, and the residue was diluted with sat. aq. NaHCO 3 (10 mL) and extracted with DCM (3 ⁇ 10 ml). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 7 2-(1H-pyrrolo[2,3-b]pyridin-5-yloxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-enyl)methyl)piperazin-1-yl)benzoic acid trifluoroacetate was prepared following the procedure described in Step 7 for Intermediate 26 using Intermediate 28-5 in place of Intermediate 26-6.
  • Step 1 A solution of t-butyl lithium (1.3 M in pentane, 60 mL, 78 mmol) was added dropwise to a solution of 1-iodo-3-methylbicyclo[1.1.1]pentane (6.5 g, 31.2 mmol) in MTBE (60 mL) at ⁇ 78° C. under N 2 . The reaction mixture was stirred for 1 h at ⁇ 78° C. Lithium 2-thienylcyanocuprate (0.25M in THF, 125 mL, 31.2 mmol) was added at ⁇ 78° C., and the addition was controlled to keep the temperature below ⁇ 60° C. After the addition, the reaction mixture was warmed to 0° C. and stirred for 30 min.
  • Step 2 To a stirred solution of benzyl 4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-ene-1-carboxylate (1.1 g, 3.39 mmol) in THF (40 mL) at 0° C. was added lithium aluminum hydride (386.6 mg, 10.2 mmol). The reaction was warmed to rt and stirred for 3 h. The reaction was then cooled to 0° C., diluted with Et 2 O (40 ml) and treated with H 2 O (0.386 mL), 0.386 mL of 15% NaOH(aq.) followed by H 2 O (1.15 mL).
  • reaction was warmed to rt, stirred for 15 min, and then treated with anhydrous MgSO 4 . After 15 min, the reaction was filtered, concentrated, and purified by column chromatography (SiO 2 , EtOAc/pet. ether) to provide (4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methanol (1.1 g, 68% yield) as a colorless oil.
  • Step 3 To a stirred solution of (4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methanol (500 mg, 2.27 mmol) in DCM (20 mL) at 0° C. was added SOCl 2 (0.537 mL, 4.54 mmol) drop wise. The reaction mixture was warmed to rt and stirred for 2 h. The reaction was concentrated, diluted with DCM and concentrated once more to obtain 1-(2-(chloromethyl)-5,5-dimethylcyclohex-1-en-1-yl)-3-methylbicyclo[1.1.1]pentane (540 mg, quantitative yield) as a clear oil.
  • Step 4 To a stirred solution of 1-(2-(chloromethyl)-5,5-dimethylcyclohex-1-en-1-yl)-3-methylbicyclo[1.1.1]pentane (540 mg, 2.26 mmol) in acetone (20 mL) was added methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(piperazin-1-yl)benzoate (798 mg, 2.26 mmol), NaI (33.90 mg, 0.22 mmol) and K 2 CO 3 (938.9 mg, 6.80 mmol) at rt. The reaction was then heated to reflux for 6 h.
  • Step 5 To a stirred solution of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate (1.15 g, 2.075 mmol) in MeOH:THF:H 2 O (1:1:1) (36 mL) was added LiOH.H 2 O (261.30 mg, 6.23 mmol) at rt. The reaction was heated to 30° C. and stirred for 16 h.
  • Step 1 A solution of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(piperazin-1-yl)benzoate (35 g, 99.3 mmol) and Intermediate 22 (26.0 g, 119.2 mmol) in THF (700 mL) was stirred at rt for 20 min. The reaction was then cooled to 0° C. and NaBH(OAc) 3 (63.15 g, 297.96 mmol) was added. Following the addition, the reaction was warmed to rt. After 16 h, the reaction was poured into ice cold water (1 L), and extracted with EtOAc (2 ⁇ 500 mL).
  • Step 2 Intermediate 28 was prepared following the procedure described in Step 5, Route B for Intermediate 28. LC/MS (ESI) m/z 541.3 [M+H] + .
  • Step 1 To a solution of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(piperazin-1-yl)benzoate (1.89 g, 5.38 mmol) in DMSO (25 mL) was added a solution of Intermediate 23 (1.5 g, 6.46 mmol) in THF (25 mL) at rt and the reaction was stirred for 1 h. The reaction was then cooled to 0° C. and treated with Na(OAc) 3 BH (3.42 g, 16.14 mmol) and warmed to rt. After 24 h, the reaction was diluted with sat. aq.
  • Step 2 Intermediate 29 was prepared following the procedure described in Step 5, Route B for Intermediate 28 using Intermediate 29-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • Step 1 Methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate (Intermediate 30-1) was prepared following the procedure described in Step 1, Route C for Intermediate 28 using Intermediate 24 in place of Intermediate 22. LCMS (ESI) m/z 591.2 [M+H] + .
  • Step 2 Intermediate 30 was prepared following the procedure described in Step 5, Route B for Intermediate 26 using Intermediate 30-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • LCMS (ESI) m/z 577.5[M+H] + .
  • Step 1 Representative procedure (reaction was performed in 3 parallel batches): To a stirred solution of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(piperazin-1-yl)benzoate (2 g, 5.68 mmol) in DMSO (0.2 M, 30 mL) was added a solution of Intermediate 25 (1.72 g, 6.22 mmol) in THF (30 mL) at rt. After 1 h, the reaction mixture was cooled to 0° C., and treated with NaBH(OAc) 3 (1.70 g, 17.04 mmol). The reaction was warmed to rt and stirred for 24 h. The reaction mixture was diluted with sat. aq.
  • Step 2 To a stirred solution of Intermediate 31-1 (8.3 g, 13.65 mmol) in MeOH:THF:H 2 O (1:1:1) (100 mL) was added LiOH.H 2 O (1.7 g, 40.95 mmol) at rt. The reaction mixture was then heated to 35° C. and stirred for 16 h. The reaction mixture was concentrated, diluted with water and neutralized with 1N HCl. The product was then extracted with 10% MeOH-DCM (3 ⁇ 150 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated to provide Intermediate 31 (7.6 g, 90% yield) as a white solid.
  • Step 1 3,3-dimethyl-1-(3-isopropylbicyclo[1.1.1]pentan-1-yl)hex-5-en-1-one (Intermediate 32-1) was prepared following the procedure described in Step 1 from Intermediate 26 using Intermediate 12 in place of Intermediate 1.
  • Step 2 E/Z-7-(3-isopropylbicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxohept-2-enenitrile (Intermediate 32-2) was prepared following the procedure described in Step 2 from Intermediate 26 using Intermediate 32-1 in place of Intermediate 26-1.
  • LC/MS (ESI) m/z 260.4 [M+H] + .
  • Step 3 7-(3-Isopropylbicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxoheptanenitrile (Intermediate 32-3) was prepared following the procedure described in Step 3 from Intermediate 26 using Intermediate 32-2 in place of Intermediate 26-2.
  • Step 4 2-(3-Isopropylbicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enecarbonitrile (Intermediate 32-4) was prepared following the procedure described in Step 4 from Intermediate 26 using Intermediate 32-3 in place of Intermediate 26-3.
  • LC/MS (ESI) m/z 244.4 [M+H] + .
  • Step 5 2-(3-Isopropylbicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enecarbaldehyde (Intermediate 32-5) was prepared following the procedure described in Step 5 from Intermediate 26 using Intermediate 32-4 in place of Intermediate 26-4.
  • LC/MS (ESI) m/z 247.4 [M+H] + .
  • Step 6 tert-Butyl 2-(1H-pyrrolo[2,3-b]pyridin-5-yloxy)-4-(4-((2-(3-isopropylbicyclo [1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enyl)methyl)piperazin-1-yl)benzoate (Intermediate 32-6) was prepared following the procedure described in Step 6, Route A for Intermediate 28 using Intermediate 32-5 in place of Intermediate 28-5. LC/MS (ESI) m/z 625.7 [M+H] + .
  • Step 7 To a solution of Intermediate 32-6 (160 mg, 0.26 mmol) in DCM (5 mL) at 0° C. was added TFA (176 mg, 1.54 mmol). The mixture was warmed to rt and stirred for 3 h. The reaction was then diluted with sat. aq. NaHCO 3 (10 mL), and extracted with DCM (3 ⁇ 10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated to provide Intermediate 32 as an off-white solid. LC/MS (ESI) m/z 569.6 [M+H] + .
  • Step 1 1-(3-(1,1-Difluoroethyl)bicyclo[1.1.1]pentan-1-yl)-3,3-dimethylhex-5-en-1-one (Intermediate 33-1) was prepared following the procedure described in Step 1 for Intermediate 26 using Intermediate 13 in place of Intermediate 1.
  • Step 2 E/Z-7-(3-(1,1-difluoroethyl)bicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxohept-2-enenitrile (Intermediate 33-2) was prepared following the procedure described in Step 2 for Intermediate 26 using Intermediate 33-1 in place of Intermediate 26-1.
  • LC/MS (ESI) m/z 282.5 [M+H] + .
  • Step 3 7-(3-(1,1-Difluoroethyl)bicyclo[1.1.1]pentan-1-yl)-5,5-dimethyl-7-oxoheptanenitrile (Intermediate 33-3) was prepared following the procedure described in Step 3 for Intermediate 26 using Intermediate 33-2 in place of Intermediate 26-2.
  • Step 4 2-(3-(1,1-Difluoroethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enecarbonitrile (Intermediate 33-4) was prepared following the procedure described in Step 4 for Intermediate 26 using Intermediate 33-3 in place of Intermediate 26-3.
  • LC/MS (ESI) m/z 266.1 [M+H] + .
  • Step 5 2-(3-(1,1-difluoroethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enecarbaldehyde (Intermediate 33-5) was prepared following the procedure described in Step 5 for Intermediate 26 using Intermediate 33-4 in place of Intermediate 26-4. LC/MS (ESI) m/z 269.5 [M+H] + .
  • Step 6 tert-butyl 2-(1H-pyrrolo[2,3-b]pyridin-5-yloxy)-4-(4-((2-(3-(1,1-difluoroethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-enyl)methyl)piperazin-1-yl)benzoate (Intermediate 33-6) was prepared following the procedure described in Step 6, Route A for Intermediate 28 using Intermediate 33-5 in place of Intermediate 28-5. LC/MS (ESI) m/z 647.3 [M+H] + .
  • Step 7 Intermediate 33 was prepared following the procedure described in Step 7 for Intermediate 32 using Intermediate 33-6 in place of Intermediate 32-6. LC/MS (ESI) m/z 591.3 [M+H] + .
  • Step 1 To a stirred solution of 3,3-dimethylpent-4-en-1-ol (18.5 g, 162.01 mmol) in DCM (100 mL), was added MsCl (13.54 mL, 175.0 mmol) followed by NEt 3 (33.87 mL, 243.0 mmol) at 0° C. and the reaction was warmed to rt. After 4 h, sat. aq. NaHCO 3 solution (100 mL) was added and the reaction was extracted with DCM (3 ⁇ 100 mL).
  • Step 2 To a pressure flask was added Intermediate 36-1 (20 g, 104.01 mmol) and NaI (46.77 g, 312.04 mmol) in acetone (100 mL). The flask was sealed and the reaction was stirred at 100° C. for 12 h. The reaction mixture was cooled to rt, diluted with water (250 mL) and extracted with Et 2 O (3 ⁇ 200 mL). The combined organic layers were washed with sat. aq. Na 2 S203, dried over Na 2 SO 4 , and evaporated to afford 5-iodo-3,3-dimethylpent-1-ene (Intermediate 36-2) (18 g, 77% yield) as a clear colorless oil.
  • Step 3 1-(3-Chlorobicyclo[1.1.1]pentan-1-yl)-4,4-dimethylhex-5-en-1-one (Intermediate 36-3) was prepared following the procedure described in Step 1 for Intermediate 26 by reacting 36-2 in place of 5-iodo-4,4-dimethylpent-1-ene.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 5.71-5.63 (m, 1H), 4.97-4.88 (m, 2H), 2.38 (s, 6H), 2.34-2.30 (m, 2H), 1.57-1.52 (m, 2H), 0.98 (s, 6H).
  • Step 4 Ozone gas was bubbled into a solution of Intermediate 36-3 (1.5 g, 6.63 mmol) in DCM (40 mL) at ⁇ 78° C. until the solution turned a blue color ( ⁇ 30 min). Then N 2 gas was bubbled into the reaction mixture until it became colorless. PPh 3 (2.6 g, 9.94 mmol) was added in one portion and the reaction was warmed to rt. After 3 h, the reaction mixture was diluted with DCM (100 mL), washed with water (2 ⁇ 25 mL), and brine (50 mL). The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 5 To a stirred solution of diethyl cyanomethylphosphonate (619 mg, 3.50 mmol) in toluene (10 mL) at 0° C. was added LiHMDS (1 M in toluene, 3.5 mL, 3.50 mmol). The reaction was then warmed to rt. After 30 min, the solution was added dropwise at ⁇ 78° C. to a solution of Intermediate 36-4 (800 mg, 3.50 mmol) in toluene (10 mL). The reaction mixture was warmed to rt and stirred for 16 h at which point it was cooled to 0° C. and quenched with sat. aq. NH 4 Cl (20 ml).
  • Step 6 A solution of Intermediate 36-5 (440 mg, 1.75 mmol) in MeOH (10 mL) was treated with Pd/C (25 wt %, 110 mg) and stirred under an atmosphere of H 2 (1 atm) for 2 h. The reaction was then purged with N 2 , and filtered over Celite. The Celite plug was washed with MeOH (3 ⁇ 25 mL) and the combined organic layers were concentrated to provide 7-(3-chlorobicyclo[1.1.1]pentan-1-yl)-4,4-dimethyl-7-oxoheptanenitrile (Intermediate 36-6) as a clear colorless oil (360 mg, 81% yield).
  • Step 7 2-(3-Chlorobicyclo[1.1.1]pentan-1-yl)-5,5-dimethylcyclohex-1-ene-1-carbonitrile (Intermediate 36-7) was prepared following the procedure described in Step 4 for Intermediate 26 by reacting Intermediate 36-6 in place of Intermediate 26-3.
  • LC/MS (ESI) m/z 236.4 [M+H] + .
  • Step 8 5,5-Dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-ene-1-carbaldehyde (Intermediate 36-8) was prepared following the procedure described in Step 5 for Intermediate 26 by reacting Intermediate 36-7 in place of Intermediate 26-4.
  • Step 9 To a stirred solution of Intermediate 36-8 (85 mg, 0.361 mmol) in EtOH (3 mL) was added tert-Butyl 4-(piperazin-1-yl)benzoate (104 mg, 0.397 mmol) and AcOH (cat.). After 15 min, the reaction was cooled to 0° C., treated with NaCNBH 3 (33.6 mg, 0.535 mmol) and warmed to rt. After 16 h, the reaction was diluted with sat. aq. NaHCO 3 and extracted with DCM (3 ⁇ 15 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 10 To a stirred solution of Intermediate 36-9 (80 mg, 0.165 mmol) in DCM (3 mL) at 0° C. was added TFA (113 mg, 0.99 mmol). The reaction was warmed to rt and stirred for 3h. The reaction was concentrated and then diluted with sat. aq. NaHCO 3 and extracted with DCM (3 ⁇ 10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated to obtain the Intermediate 36 as an off-white solid (60 mg, 85%). LC/MS (ESI) m/z 429.5 [M+H] + .
  • Step 1 To a stirred solution of (R)-3-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-4-(phenylthio)butanoic acid (6.8 g, 15.7 mmol) in DCM (70 mL) and DMF (10 mL) was added HATU (9.5 g, 25.12 mmol) followed by DIPEA (8.3 mL, 47.1 mmol) at 0° C. After 10 min, 4-hydroxypiperidine (2.4 g, 23.55 mmol) was added and temperature was raised to rt. After 16 h, the reaction was diluted with water and extracted with EtOAc.
  • Step 2 To a stirred solution of Intermediate 37-1 (2.75 g, 5.32 mmol) in CH 3 CN (20 mL) at rt was added diethylamine (3.3 mL, 31.92 mmol) and stirred at rt. After 16 h, the reaction was concentrated and purified by column chromatography (neutral alumina, MeOH/DCM) to afford (R)-3-amino-1-(4-hydroxypiperidin-1-yl)-4-(phenylthio)butan-1-one (Intermediate 37-2) (900 mg, 57% yield) as a brown liquid. LC/MS (ESI) m/z 295.1 [M+H] + .
  • Step 3 To a stirred solution of Intermediate 37-2 (0.9 g, 3.06 mmol) in anhydrous THF (12 mL) at 0° C. was added BH 3 (1 M in THF, 9.18 mL, 9.18 mmol) and the temperature was raised to 45° C. After 16 h, the reaction was cooled to 0° C. and MeOH (30 ml) was added. After 1 hour, the reaction was concentrated and purified by column chromatography (C18, CH 3 CN/Water) to afford (R)-1-(3-amino-4-(phenylthio)butyl)piperidin-4-ol (Intermediate 37-3) (305 mg, 36% yield) as an off-white semi solid. LC/MS (ESI) m/z 281.2 [M+H] + .
  • Step 4 To a stirred solution of Intermediate 37-3 (100 mg, 0.357 mmol) in DMF (1 mL) was added 4-fluoro-3-(trifluoromethylsulfonyl)benzenesulfonamide (99 mg, 0.32 mmol) followed by DIPEA (140 mg, 1.07 mmol) and the resulting reaction mixture was stirred at rt. After 16 h, the reaction was concentrated, diluted with water and extracted with 9:1 DCM:MeOH (2 ⁇ 10 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by trituration with EtOAc/Et 2 O to afford Intermediate 37 (105 mg, 51% yield) as a white solid. LC/MS (ESI) m/z 568.1 [M+H] + .
  • Step 1 4,4-Dimethyl-1-(3-methylbicyclo[1.1.1]pentan-1-yl)hex-5-en-1-one (Intermediate 38-1) was prepared following the procedure described in Step 1 for Intermediate 26 using Intermediate 10 and Intermediate 36-2 in place of Intermediate 1 and 5-iodo-4,4-dimethylpent-1-ene.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 5.73-5.66 (m, 1H), 4.95-4.88 (m, 2H), 2.33-2.28 (m, 2H), 1.88 (s, 6H), 1.55-1.51 (m, 2H), 1.21 (s, 3H), 0.99 (s, 6H).
  • Step 2 2,2-dimethyl-5-(3-methylbicyclo[1.1.1]pentan-1-yl)-5-oxopentanal (Intermediate 38-2) was prepared following the procedure described in Step 4 for Intermediate 36 using Intermediate 38-1 in place of Intermediate 36-3.
  • 1H NMR 300 MHz, CDCl 3 ) ⁇ 9.41 (s, 1H), 2.36-2.30 (m, 2H), 1.88 (s, 6H), 1.79-1.71 (m, 2H), 1.18 (s, 3H), 1.05 (s, 6H).
  • Step 3 4,4-dimethyl-7-(3-methylbicyclo[1.1.1]pentan-1-yl)-7-oxohept-2-enenitrile (Intermediate 38-3) was prepared following the procedure described in Step 5 for Intermediate 36 using Intermediate 38-2 in place of Intermediate 36-4.
  • LC/MS (ESI) m/z 232.5 [M+H] + .
  • Step 4 4,4-dimethyl-7-(3-methylbicyclo[1.1.1]pentan-1-yl)-7-oxoheptanenitrile (Intermediate 38-4) was prepared following the procedure described in Step 6 for Intermediate 36 using Intermediate 38-3 in place of Intermediate 36-5.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 2.38-2.33 (m, 2H), 2.29-2.25 (m, 2H), 1.90 (s, 6H), 1.62-1.58 (m, 2H), 1.48-1.44 (m, 2H), 1.19 (s, 3H), 0.90 (s, 6H).
  • Step 5 5,5-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-ene-1-carbonitrile (Intermediate 38-5) was prepared following the procedure described in Step 4 for Intermediate 26 using Intermediate 38-4 in place of Intermediate 26-3.
  • Step 6 5,5-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-ene-1-carbaldehyde (Intermediate 38-6) was prepared following the procedure described in Step 5 for Intermediate 26 using Intermediate 38-5 in place of Intermediate 26-4.
  • Step 7 tert-Butyl 4-(4-((5,5-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate (Intermediate 38-7) was prepared following the procedure described in Step 9 from Intermediate 36 using Intermediate 38-6 in place of Intermediate 36-8. LC/MS (ESI) m/z 465.6 [M+H] + .
  • Step 8 Intermediate 38 was prepared following the procedure described in Step 10 from Intermediate 36 by reacting Intermediate 38-7 in place of Intermediate 36-9.
  • LC/MS (ESI) m/z 409.6 [M+H] + .
  • Step 1 To a stirred solution of methyl 4-(piperazin-1-yl)benzoate (1.68 g, 7.6 mmol) and Intermediate 22 (2.0 g, 9.15 mmol) in THF (20 mL) was added Na(OAc) 3 BH (4.8 g, 22.8 mmol) at rt. After 16 h, the reaction was put in an ice batch and quenched with sat. aq. NaHCO 3 (25 mL). The reaction mixture was extracted with EtOAc (3 ⁇ 50 mL), dried over Na 2 SO 4 , filtered, and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 2 Intermediate 39 was prepared following the procedure described in Step 5, Route B for Intermediate 28 using Intermediate 39-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • Step 1 Methyl 4-(4-((2-(3-ethylbicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate (Intermediate 40-1) was prepared following the procedure described in Step 1 for Intermediate 39 using Intermediate 23 in place of Intermediate 22. LC/MS (ESI) m/z 437.3 [M+H] + .
  • Step 1 Intermediate 40 was prepared following the procedure described in Step 2 for Intermediate 39 using Intermediate 40-1 in place of Intermediate 39-1.
  • LC/MS (ESI) m/z 423.3 [M+H] + .
  • Step 1 To a stirred solution of Intermediate 25 (3.5 g, 12.85 mmol) in toluene was added titanium (IV) ethoxide (3.73 g, 16.36 mmol). After 30 min, a solution of methyl 4-(piperazin-1-yl) benzoate (2.35 g, 10.71 mmol) in toluene (20 mL) was added and the resulting reaction mixture was stirred at rt for 1 h. The reaction mixture was then cooled to 0° C., and Na(OAc) 3 BH (6.9 g, 32.72 mmol) was added and the reaction was warmed to rt.
  • Step 2 Intermediate 41 was prepared following the procedure described in Step 2 for Intermediate 39 by reacting Intermediate 41-1 in place of Intermediate 39-1.
  • LC/MS (ESI) m/z 463.2 [M+H] + .
  • Step 1 Methyl 4-(4-((2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate (Intermediate 42-1) was prepared following the procedure described in Step 1 for Intermediate 39 using Intermediate 24 in place of Intermediate 22.
  • Step 1 Intermediate 42 was prepared following the procedure described in Step 2 for Intermediate 39 using Intermediate 42-1 in place of Intermediate 39-1.
  • LC/MS (ESI) m/z 445.6 [M+H] + .
  • Step 1 To a stirred solution of N,N-dimethylpiperidin-4-amine (462.5 mg, 3.61 mmol), DMAP (367.80 mg, 3.01 mmol), and EDC.HCl (863.75 mg, 4.51 mmol) in DCM (20 mL) was added (R)-4-(phenylthio)-3-((4-sulfamoyl-2-((trifluoromethyl)sulfonyl)phenyl)amino)butanoic acid (prepared following a procedure described in WO2012017251A1) (1.5 g, 3.01 mmol) and Et 3 N (0.84 mL, 6.02 mmol) at rt.
  • reaction was heated to 35° C. and stirred for 16 h.
  • the reaction mixture was then cooled to rt, diluted with DCM (100 mL) and MeOH (10 mL) and washed with 10% CH 3 CO 2 H (aq.) (2 ⁇ 20 mL).
  • the organic layer was then washed with 5% NaHCO 3 (aq.) (20 mL) and 5% NaCl(aq.) (20 mL) and concentrated.
  • Step 2 To a stirred solution of Intermediate 45-1 (800 mg, 1.31 mmol) in THF (15 mL) was added BH 3 .THF (1M in THF, 6.57 mL, 6.57 mmol) at rt. The resulting reaction mixture was heated to 55° C. for 24 h in a sealed tube. The reaction was then cooled to rt, and treated with MeOH (8 mL) and conc. HCl (2 mL) and heated to 65° C. After 10 h. the reaction was concentrated, diluted with 2N NaOH solution and extracted with EtOAc. The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (C18, CH 3 CN/H 2 O) to afford Intermediate 45 (490 mg, 62% yield). LC/MS (ESI) m/z 595.3[M+H] + .
  • Step 1 (R)-tert-Butyl 4-(4-(phenylthio)-3-((4-sulfamoyl-2-((trifluoromethyl)sulfonyl)phenyl)-amino)butanoyl)piperazine-1-carboxylate (Intermediate 46-1) was prepared following the procedure described in Step 1 for Intermediate 45 using tert-butyl piperazine-1-carboxylate in place of N,N-dimethylpiperidin-4-amine. LC/MS (ESI) m/z 665.4 [M ⁇ H] ⁇ .
  • Step 2 Intermediate 46 was prepared following the procedure described in Step 2 for Intermediate 45 using Intermediate 46-1 in place of Intermediate 45-1.
  • LC/MS (ESI) m/z 653.2 [M+H] + .
  • Step 1 To a stirred solution of Intermediate 24-2 (4.67 g, 19.15 mmol) in Et 2 O (30 mL) under argon was added sec-BuLi (1.4 M in cyclohexane, 20.8 mL, 29.12 mmol) at ⁇ 78° C. and the reaction was stirred for 10 minutes at the same temperature. The temperature was then warmed to 0° C. and stirred for 1 h. The reaction was cooled to ⁇ 78° C. and a solution of Intermediate 47 (2 g, 8.32 mmol) in Et 2 O (20 mL) was added dropwise for 5 minutes. The reaction was stirred at ⁇ 78° C. for 1 h, and then warmed to 0° C. and stirred for 1 h.
  • reaction mixture was quenched with sat. aq. NH 4 Cl solution (50 mL) at 0° C., and extracted with Et 2 O (3 ⁇ 150 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated to provide 7-(diethoxymethyl)-6-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)spiro[3.5]nonan-6-ol(Intermediate 48-1)(1.5 g, crude) as a yellow oil. This was used in the next step without further purification.
  • Step 2 To a stirred solution of Intermediate 48-1 (1.5 g crude, 4.18 mmol) in 1,4-dioxane (30 mL) was added 2N HCl (aq.) (7 mL) and the resulting reaction mixture was stirred at 65-70° C. for 16 h. The reaction mixture was diluted with ice cold water (15 mL) and extracted with Et 2 O (3 ⁇ 100 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated. The product was purified by column chromatography (SiO 2 , Et 2 O/pet. ether) to afford Intermediate 48 (1 g, 45% yield over 2 steps) as a brown oil.
  • Step 1 7-(diethoxymethyl)-6-(3-methylbicyclo[1.1.1]pentan-1-yl)spiro[3.5]nonan-6-ol (Intermediate 49-1) was prepared following the procedure described in Step 1 for Intermediate 48 using 1-iodo-3-methylbicyclo[1.1.1]pentane in place of Intermediate 24-2.
  • Step 2 Intermediate 49 was prepared following the procedure described in Step 2 for Intermediate 49 using Intermediate 49-1 in place of Intermediate 48-1.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 10.23 (s, 1H), 2.23-2.20 (m, 2H), 1.96 (s, 6H), 1.89-1.71 (m, 8H), 1.58-1.55 (m, 2H), 1.16 (s, 3H).
  • Step 1 Methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((6-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)spiro[3.5]non-6-en-7-yl)methyl)piperazin-1-yl)benzoate (Intermediate 50-1) was prepared following the procedure described in Step 1, Route C for Intermediate 28 using Intermediate 48 in place of Intermediate 22. LCMS (ESI) m/z 603.5 [M+H] + .
  • Step 2 Intermediate 50 was prepared following the procedure described in Step 5, Route B for Intermediate 28 using Intermediate 50-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • LC/MS (ESI) m/z 589.3.
  • Step 1 Methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((6-(3-methylbicyclo[1.1.1]pentan-1-yl)spiro[3.5]non-6-en-7-yl)methyl)piperazin-1-yl)benzoate: (Intermediate 51-1) was prepared following the procedure described in Step 1, Route C for Intermediate 28 using Intermediate 49 in place of Intermediate 22. LC/MS (ESI) m/z 567.3 [M+H] + .
  • Step 2 Intermediate 50 was prepared following the procedure described in Step 5, Route B for Intermediate 28 using Intermediate 51-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • Step 1 2-(diethoxymethyl)-1-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohexanol (Intermediate 56-1) was prepared following the procedure described in Step 1, for Intermediate 25 using Intermediate 24-2 in place of i-iodo-3(trifluoromethyl)bicyclo[1.1.1]pentane and 2-(diethoxymethyl)-4,4-dimethylcyclohexanone in place of Intermediate 19. The crude product was used in the next step without purification.
  • Step 2 2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-5,5-dimethylcyclohex-1-enecarbaldehyde (Intermediate 56-2) was prepared following the procedure described in Step 2 for Intermediate 22, using Intermediate 56-1 in place of Intermediate 22-1.
  • Step 3 To a stirred solution of methyl 4-(piperazin-1-yl)benzoate (389 mg, 1.77 mmol) in THF (10 ml) was added a solution of Intermediate 56-2 (450 mg, 1.77 mmol) in THF (5 mL) at rt. The reaction was stirred for 1h, treated with Na(OAc) 3 BH (1.12 g, 5.31 mmol) at 0° C., and then warmed to rt. After 16 h, MeOH (10 mL) was added and the reaction was stirred for 30 minutes. The reaction mixture was concentrated under reduced pressure, dissolved in DCM (20 mL) and washed with sat. aq. NaHCO 3 (3 ⁇ 10 mL).
  • Step 4 Intermediate 56 was prepared following the procedure described in Step 5, Route B for Intermediate 28 using Intermediate 56-3 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • LC/MS (ESI) m/z 445.4 [M+H] + .
  • Step 1 To a solution of (R)-4-(phenylthio)-3-((4-sulfamoyl-2-((trifluoromethyl)sulfonyl)phenyl)amino)butanoic acid (1.5 g, 3.01 mmol) and 0-(Benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate (TBTU) (1.09 g, 3.41 mmol) in DCM (3 mL) at 0° C. was added N-methylmorpholine (1.3 mL, 9.3 mmol) and DMF (1.5 mL). The reaction was warmed to rt and stirred for 0.5 h.
  • TBTU 0-(Benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate
  • reaction mixture was then cooled to 0° C., and azetidin-3-ol (264 mg, 3.61 mmol) was added and the reaction was warmed to rt. After 16h, the reaction was quenched with sat. aq. NaHCO 3 (50 mL) and extracted with EtOAc (3 ⁇ 100 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated.
  • Step 2 To a stirred solution of Intermediate 57-1 (1.0 g, 1.80 mmol) in THF (20 mL) at 0° C. was added BH 3 .THF (1 M in THF, 5.0 mL, 5 mmol) and the reaction was warmed to rt. After 1 h, the reaction mixture was heated to 55° C. and stirred for 16 h in a sealed tube. The reaction mixture was then cooled to 0° C., quenched with NH 3 (7.0 M in MeOH, 5 mL) at 0° C. and warmed to rt. After 16 h.
  • Step 1 Methyl 4-(4-((6-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)spiro[3.5]non-6-en-7-yl)methyl)piperazin-1-yl)benzoate(Intermediate 58-1) was prepared following the procedure described in Step 3, for Intermediate 56 using Intermediate 48 in place of Intermediate 56-2. LC/MS (ESI) m/z 471.3 [M+H] + .
  • Step 2 Intermediate 58 was prepared following the procedure described in Step 5, Route B for Intermediate 28 using Intermediate 58-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate.
  • LC/MS (ESI) m/z 457.5 [M+H] + .
  • Step 1 (R)-4-((4-(4-(2-((tert-butyldiphenylsilyl)oxy)ethyl)piperazin-1-yl)-4-oxo-1-(phenylthio)butan-2-yl)amino)-3-((trifluoromethyl)sulfonyl)benzenesulfonamide (Intermediate 59-1) was prepared following the procedure described in Step 1 for Intermediate 45 using 1-(2-((tert-butyldiphenylsilyl)oxy)ethyl)piperazine in place of N,N-dimethylpiperidin-4-amine. LC/MS (ESI) m/z 849.3 [M+H] + .
  • Step 2 Intermediate 59 was prepared following the procedure described in Step 2, for Intermediate 57 using Intermediate 59-1 in place of Intermediate 57-1.
  • LC/MS (ESI) m/z 835.0 [M+H] + .
  • Step 1 2-((tert-butyldiphenylsilyl)oxy)-N-ethylethanamine (Intermediate 60-1) was prepared following a procedure described in WO2012/017251A1. LC/MS (ESI) m/z 328.4 [M+H] + .
  • Step 2 To a stirred solution of (R)-4-(phenylthio)-3-((4-sulfamoyl-2-((trifluoromethyl)sulfonyl)phenyl)amino)butanoic acid (500 mg, 1.0 mmol) in CH 3 CN (10 mL) at 0° C. was added Intermediate 60-1 (328 mg, 1.01 mmol) in CH 3 CN (2 mL), followed by N-methyl imidazole (250 mg, 3.1 mmol) and N,N,N′,N′-tetramethylchloroformamidinium hexafluorophosphate (TCFH) (308 mg, 1.1 mmol).
  • TCFH N,N,N′,N′-tetramethylchloroformamidinium hexafluorophosphate
  • the reaction was warmed to rt and stirred for 16 h. The reaction was then diluted with water and extracted with EtOAc (3 ⁇ 100 mL). The combined organic layers were washed with sat. aq. NaHCO 3 (2 ⁇ 20 mL), water (2 ⁇ 10 mL) and then brine (2 ⁇ 20 mL). The organic layer was dried over Na 2 SO 4 , filtered and concentrated. The crude product was purified by column chromatography (SiO 2 , EtOAc/pet.
  • Step 2 Intermediate 60 was prepared following the procedure described in Step 2, for Intermediate 57 using Intermediate 60-2 in place of Intermediate 57-1.
  • LC/MS (ESI) m/z 794.8 [M+H] + .
  • Step 1 4-(((2R)-4-(3-Hydroxypyrrolidin-1-yl)-4-oxo-1-(phenylthio)butan-2-yl)amino)-3-((trifluoromethyl)sulfonyl)benzenesulfonamide (Intermediate 61-1) was prepared following the procedure described in Step 1, for Intermediate 45 using pyrrolidin-3-ol in place of N,N-dimethylpiperidin-4-amine. LCMS (ESI) m/z 568.1 [M+H] + .
  • Step 2 Intermediate 61 was prepared following the procedure described in Step 2, for Intermediate 57 using Intermediate 61-1 in place of Intermediate 57-1.
  • Step 1 1-(3-Chlorobicyclo[1.1.1]pentan-1-yl)hex-5-en-1-one (Intermediate 62-1) was prepared following the procedure described in Step 1 for Intermediate 26 using 5-bromopent-1-ene in place of 5-iodo-3,3-dimethylpent-1-ene.
  • Step 2 E/Z-7-(3-chlorobicyclo[1.1.1]pentan-1-yl)-7-oxohept-2-enenitrile (Intermediate 62-2) was prepared following the procedure described in Step 2 for Intermediate 26 using Intermediate 62-1 in place of Intermediate 26-1.
  • LC/MS (ESI) m/z 236.3 [M+H] + .
  • Step 3 7-(3-Chlorobicyclo[1.1.1]pentan-1-yl)-7-oxoheptanenitrile (Intermediate 62-3) was prepared following the procedure described in Step 3 for Intermediate 26 using Intermediate 62-2 in place of Intermediate 26-2.
  • Step 4 2-(3-chlorobicyclo[1.1.1]pentan-1-yl)cyclohex-1-enecarbonitrile (Intermediate 62-4) was prepared following the procedure described in Step 4 for Intermediate 26 using Intermediate 62-3 in place of Intermediate 26-3.
  • LC/MS (ESI) m/z 208.1 [M+H] + .
  • Step 5 2-(3-chlorobicyclo[1.1.1]pentan-1-yl)cyclohex-1-enecarbaldehyde (Intermediate 62-5) was prepared following the procedure described in Step 5 for Intermediate 26 using Intermediate 62-4 in place of Intermediate 26-4.
  • 1 H NMR 300 MHz, CDCl 3 ) ⁇ 10.16 (s, 1H), 2.46 (s, 6H), 2.23-2.21 (m, 2H), 2.15-2.13 (m, 2H), 1.64-1.54 (m, 4H).
  • Step 6 tert-butyl 2-(1H-pyrrolo[2,3-b]pyridin-5-yloxy)-4-(4-((2-(3-chlorobicyclo[1.1.1]pentan-1-yl)cyclohex-1-enyl)methyl)piperazin-1-yl)benzoate (Intermediate 62-6) was prepared following the procedure described in Step 6, Route A for Intermediate 28 using Intermediate 62-5 in place of Intermediate 22. LC/MS (ESI) m/z 589.3 [M+H] + .
  • Step 7 Intermediate 62 was prepared following the procedure described in Step 7, for Intermediate 32, using Intermediate 62-6 in place of Intermediate 32-6.
  • LC/MS (ESI) m/z 533.3 [M+H] + .
  • N,N-dimethylethylenediamine (2-2.5 equiv., Note #2) was added to the reaction mixture and the reaction was stirred for 90 min.
  • the reaction mixture was then washed with 10% aq. AcOH (Note #3), 5% NaHCO 3 (aq.) and then with 5% NaCl (aq.).
  • the organic layer was concentrated, and crude product C was either purified by 1) column chromatography (SiO 2 ), 2) HPLC (10 mM NH 4 CO 3 H(aq): CH 3 CN or MeOH), or 3) trituration with an organic solvent.
  • Example 1 (69 mg, 39%) as a yellow solid.
  • Example 2 was prepared following General Procedure A using Intermediate 26 and Intermediate 3.
  • Example 3 was prepared following General Procedure A using Intermediate 26 and Intermediate 4.
  • Example 4 was prepared following General Procedure A using Intermediate 26 and Intermediate 5.
  • Example 5 was prepared following General Procedure A using Intermediate 26 and Intermediate 6.
  • Example 6 was prepared following General Procedure A using Intermediate 26 and Intermediate 7. LC/MS (ESI) m/z 857.4 [M+H] + .
  • Example 7 was prepared following General Procedure A using Intermediate 26 and Intermediate 8. LC/MS (ESI) m/z 886.5 [M+H] + .
  • Example 8 was prepared following General Procedure A using Intermediate 26 and Intermediate 9. LC/MS (ESI) m/z 871.6 [M+H] + .
  • Example 9 was prepared following General Procedure A using Intermediate 28 and 3-nitro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)benzenesulfonamide.
  • H NMR 400 MHz, DMSO-d 6 ) ⁇ 11.68 (s, 1H), 11.42 (br s, 1H), 8.58 (br s, 1H), 8.54 (s, 1H), 8.03 (s, 1H), 7.80-7.76 (m, 1H), 7.53-7.48 (m, 3H), 7.12-7.05 (m, 1H), 6.74-6.70 (m, 1H), 6.38 (s, 1H), 6.25 (s, 1H), 3.85-3.83 (m, 2H), 3.30-3.23 (m, 4H), 3.19-3.05 (m, 4H), 2.99 (br s, 2H), 2.38-2.32 (m, 4H), 2.05-1.95 (m, 2H), 1.93-1.85 (m, 1H), 1.75 (
  • Example 10 was prepared following General Procedure A using Intermediate 27 and 3-nitro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)benzenesulfonamide.
  • Example 11 was prepared following General Procedure A using Intermediate 28 and Intermediate 6.
  • Example 13 was prepared following General Procedure A using Intermediate 28 and Intermediate 15.
  • Example 14 was prepared following General Procedure A using Intermediate 28 and Intermediate 16. LC/MS (ESI) m/z 865.5 [M+H] + .
  • Example 15 was prepared following General Procedure A using Intermediate 28 and Intermediate 52.
  • Example 16 was prepared following General Procedure A using Intermediate 28 and Intermediate 53.
  • Example 17 was prepare following General Procedure A using Intermediate 28 and Intermediate 17.
  • Example 18 was prepared following General Procedure A using Intermediate 28 and Intermediate 17A.
  • LCMS (ESI) m/z 853.5[M+H] + .
  • the absolute stereochemistry was arbitrarily assigned.
  • Example 19 was prepared following General Procedure A using Intermediate 28 and Intermediate 17B. LC/MS (ESI) m/z 853.6[M+H] + . The absolute stereochemistry was arbitrarily assigned.
  • Example 19 was prepared following General Procedure A using Intermediate 28 and Intermediate 18. LC/MS (ESI) m/z 866.5 [M+H] +
  • Example 21 was prepared following General Procedure A using Intermediate 28 and Intermediate 54. LC/MS (ESI) m/z 853.6 [M+H] + .
  • N,N-dimethylethylenediamine (0.055 mL) was charged to the reaction mixture, and stirring was continued for 90 min. The reaction mixture was then washed with 10% acetic acid solution (2 ⁇ 10 mL). Note: The organic layer was diluted with DCM (10 mL) and MeOH (3 mL), before final separation of the aqueous layer. The organic layer was washed with 5% NaHCO 3 (aq.) (10 mL), 5% NaCl(aq.) (10 mL) and the organic layer was concentrated.
  • Example 23 was prepared following General Procedure B using Intermediate 29 and 3-nitro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)benzenesulfonamide.
  • Example 24 was prepared following General Procedure B using Intermediate 29 and Intermediate 15. L/MS (ESI) m/z 883.9 [M+H] + .
  • Example 25 was prepared following General Procedure B using Intermediate 29 and Intermediate 52. LC/MS (ESI) m/z 870.8 [M+H] + .
  • Example 26 was prepared following General Procedure B using Intermediate 29 and Intermediate 53. LC/MS (ESI) m/z 871.7 [M+H] + .
  • Example 27 was prepared following General Procedure B using Intermediate 29 and Intermediate 18. LC/MS (ESI) m/z 880.6 [M+H]+.
  • Example 28 was prepared following General Procedure A using Intermediate 31 and 3-nitro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)benzenesulfonamide.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • AIDS & HIV (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Pyrrole Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Pyridine Compounds (AREA)
  • Nitrogen- Or Sulfur-Containing Heterocyclic Ring Compounds With Rings Of Six Or More Members (AREA)
  • Cephalosporin Compounds (AREA)
US16/957,862 2018-01-10 2019-01-08 Benzamide compounds Active US11344546B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/957,862 US11344546B2 (en) 2018-01-10 2019-01-08 Benzamide compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862615857P 2018-01-10 2018-01-10
PCT/US2019/012704 WO2019139902A1 (fr) 2018-01-10 2019-01-08 Composés benzamide
US16/957,862 US11344546B2 (en) 2018-01-10 2019-01-08 Benzamide compounds

Publications (2)

Publication Number Publication Date
US20210009543A1 US20210009543A1 (en) 2021-01-14
US11344546B2 true US11344546B2 (en) 2022-05-31

Family

ID=67218673

Family Applications (6)

Application Number Title Priority Date Filing Date
US16/957,861 Active US11318134B2 (en) 2018-01-10 2019-01-08 Benzamide compounds
US16/957,865 Active 2039-08-27 US11590126B2 (en) 2018-01-10 2019-01-08 Benzamide compounds
US16/957,862 Active US11344546B2 (en) 2018-01-10 2019-01-08 Benzamide compounds
US17/656,726 Active US11813259B2 (en) 2018-01-10 2022-03-28 Benzamide compounds
US18/202,622 Active US11813260B1 (en) 2018-01-10 2023-05-26 Benzamide compounds
US18/471,017 Pending US20240180899A1 (en) 2018-01-10 2023-09-20 Benzamide compounds

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US16/957,861 Active US11318134B2 (en) 2018-01-10 2019-01-08 Benzamide compounds
US16/957,865 Active 2039-08-27 US11590126B2 (en) 2018-01-10 2019-01-08 Benzamide compounds

Family Applications After (3)

Application Number Title Priority Date Filing Date
US17/656,726 Active US11813259B2 (en) 2018-01-10 2022-03-28 Benzamide compounds
US18/202,622 Active US11813260B1 (en) 2018-01-10 2023-05-26 Benzamide compounds
US18/471,017 Pending US20240180899A1 (en) 2018-01-10 2023-09-20 Benzamide compounds

Country Status (15)

Country Link
US (6) US11318134B2 (fr)
EP (3) EP3737681A4 (fr)
JP (4) JP7355741B2 (fr)
KR (3) KR20200108298A (fr)
CN (5) CN118084904A (fr)
AR (4) AR114998A1 (fr)
AU (3) AU2019207616A1 (fr)
BR (3) BR112020014160A2 (fr)
CA (3) CA3087263A1 (fr)
IL (3) IL275761A (fr)
MX (4) MX2020007388A (fr)
RU (2) RU2020120929A (fr)
SG (3) SG11202005785XA (fr)
TW (4) TW201934550A (fr)
WO (4) WO2019139900A1 (fr)

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110177788B (zh) 2017-01-07 2023-03-24 重庆复创医药研究有限公司 作为bcl-2选择性凋亡诱导剂的化合物
PL3612531T3 (pl) 2017-04-18 2022-12-19 Shanghai Fochon Pharmaceutical Co., Ltd. Środki wywołujące apoptozę
CN118084904A (zh) 2018-01-10 2024-05-28 里科瑞尔姆Ip控股有限责任公司 苯甲酰胺化合物
KR20220034038A (ko) * 2019-07-10 2022-03-17 리커리엄 아이피 홀딩스, 엘엘씨 Bcl-2 억제제의 나노입자 제형
JP2022540333A (ja) * 2019-07-10 2022-09-15 リキュリウム アイピー ホールディングス リミテッド ライアビリティー カンパニー Bcl-2タンパク質阻害剤
CN115175677A (zh) 2019-12-06 2022-10-11 罗索肿瘤学公司 布鲁顿氏酪氨酸激酶抑制剂的给药
CA3165474A1 (fr) * 2019-12-20 2021-06-24 Recurium Ip Holdings, Llc Associations
US20230008362A1 (en) * 2019-12-20 2023-01-12 Recurium Ip Holdings, Llc Combinations
CN115023229A (zh) * 2019-12-20 2022-09-06 里科瑞尔姆Ip控股有限责任公司 组合
IL294067A (en) * 2019-12-20 2022-08-01 Recurium Ip Holdings Llc conjunctions
WO2021167987A1 (fr) * 2020-02-21 2021-08-26 Recurium Ip Holdings, Llc Composés difluorométhyl iodo et procédés
MX2022010512A (es) * 2020-04-28 2022-11-16 Recurium Ip Holdings Llc Inhibidores de proteína bcl-2.
US20230158048A1 (en) * 2020-05-07 2023-05-25 Recurium Ip Holdings, Llc Combinations
IL298297A (en) * 2020-05-19 2023-01-01 Recurium Ip Holdings Llc Treatment of amyloidosis
EP4228645A1 (fr) * 2020-10-16 2023-08-23 Recurium IP Holdings, LLC Combinaisons d'inhibiteurs de bcl-2 avec des agents chimiothérapeutiques
WO2022133446A1 (fr) * 2020-12-16 2022-06-23 Recurium Ip Holdings, Llc Combinaisons
CN113143930B (zh) * 2021-04-08 2023-05-30 深圳湾实验室 化合物在制备SARS-Cov-2 E蛋白抑制剂中的用途
CN113816889B (zh) * 2021-10-11 2023-08-22 中国药科大学 一类4-磺酰亚胺-1H-吡咯类Mcl-1抑制剂及其抗肿瘤医药用途

Citations (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003096461A (ja) 2001-09-25 2003-04-03 Fuji Photo Film Co Ltd 液晶組成物およびそれを用いた液晶素子
WO2003089441A1 (fr) 2002-04-22 2003-10-30 Ibfb Gmbh Mercaptans tricycliques, leur procede de production et leur utilisation
WO2005049593A2 (fr) 2003-11-13 2005-06-02 Abbott Laboratories Promoteurs de l'apoptose contenant n-acylsulfonamide
WO2005079803A1 (fr) 2004-02-13 2005-09-01 Pfizer Products, Inc. Composes pour traitement des maladies cardio-vasculaires
US20060128706A1 (en) 2003-11-13 2006-06-15 Milan Bruncko Apoptosis promoters
WO2006093346A1 (fr) 2005-03-03 2006-09-08 Fujifilm Corporation Film d’acylate de cellulose, plaque polarisante et dispositif d’affichage a cristaux liquides
US20060258657A1 (en) 2003-11-13 2006-11-16 Milan Bruncko Apoptosis promoters
US20070015787A1 (en) 2003-11-13 2007-01-18 Milan Bruncko Apoptosis promoters
US20070027135A1 (en) 2005-05-12 2007-02-01 Milan Bruncko Apoptosis promoters
WO2008008374A2 (fr) 2006-07-14 2008-01-17 Chemocentryx, Inc. Inhibiteurs de ccr2 et leurs procédés d'utilisation
WO2008030836A2 (fr) 2006-09-05 2008-03-13 Abbott Laboratories Traitement des maladies myéloprolifératives
WO2008033747A2 (fr) 2006-09-11 2008-03-20 Curis, Inc. Petites molécules multifonctionnelles servant d'agents anti-prolifératifs
WO2008064116A2 (fr) 2006-11-16 2008-05-29 Abbott Laboratories Procédé pour éviter ou traiter les rejets lors de transplantations d'organes, de cellules souches hématopoïétiques ou de moelle osseuse
US20080199873A1 (en) 2006-12-04 2008-08-21 Anderson Mark G Companion diagnostic assays for cancer therapy
WO2009036051A1 (fr) 2007-09-10 2009-03-19 Curis, Inc. Inhibiteurs de la bcl-2 contenant une fraction de liaison au zinc
WO2010065824A2 (fr) 2008-12-04 2010-06-10 Abbott Laboratories Agents induisant l'apoptose pour le traitement du cancer et de maladies immunes et auto-immunes
WO2010065865A2 (fr) 2008-12-05 2010-06-10 Abbott Laboratories Agents induisant l'apoptose sélectifs pour bcl-2 pour le traitement du cancer et des maladies immunes
US20100160322A1 (en) 2008-12-04 2010-06-24 Abbott Laboratories Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2010083442A1 (fr) 2009-01-19 2010-07-22 Abbott Laboratories Agents induisant une apoptose pour le traitement du cancer et de maladies immunes et auto-immunes
CN101798292A (zh) 2010-03-29 2010-08-11 无锡好芳德药业有限公司 ABT-263衍生的新型Bcl-2蛋白抑制剂的制备
US20100227838A1 (en) 2005-05-12 2010-09-09 Abbott Laboratories Combination Therapy for Treating Cancer and Diagnostic Assays for Use Therein
US20100305122A1 (en) 2009-05-26 2010-12-02 Abbott Laboratories Apoptosis inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2010138588A2 (fr) 2009-05-26 2010-12-02 Abbott Laboratories Agents induisant l'apoptose, dans le traitement du cancer et de maladies immunes et auto-immunes
US20100311751A1 (en) 2009-06-08 2010-12-09 Abbott Laboratories Solid dispersions containing an apoptosis-promoting agent
US20100323020A1 (en) 2009-06-18 2010-12-23 Abbott Laboratories Stable nanoparticulate drug suspension
WO2010148422A1 (fr) 2009-06-25 2010-12-29 Walter And Eliza Hall Institute Of Medical Research Composés et procédés de traitement d'infestations parasitaires
WO2011021038A1 (fr) 2009-08-20 2011-02-24 Karus Therapeutics Limited Composés hétérocycliques tricycliques en tant qu'inhibiteurs de phosphoinositide 3-kinase
WO2011150016A1 (fr) 2010-05-26 2011-12-01 Abbott Laboratories Agents inducteurs d'apoptose pour le traitement du cancer et de maladies immunitaires et auto-immunes
WO2012058392A1 (fr) 2010-10-29 2012-05-03 Abbott Laboratories Dispersions solides contenant un agent induisant l'apoptose
US20120108590A1 (en) 2010-10-29 2012-05-03 Abbott Laboratories Melt-extruded solid dispersions containing an apoptosis-inducing agent
WO2012063085A2 (fr) 2010-11-11 2012-05-18 Redx Pharma Limited Dérivés de médicaments
US20120129853A1 (en) 2010-11-23 2012-05-24 Abbott Laboratories Methods of treatment using selective bcl-2 inhibitors
US20120189539A1 (en) 2011-01-25 2012-07-26 The Regents Of University Of Michigan Bcl-2/bcl-xl inhibitors and therapeutic methods using the same
US8557983B2 (en) 2008-12-04 2013-10-15 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2013185202A1 (fr) 2012-06-14 2013-12-19 Beta Pharma Canada Inc Inducteurs d'apoptose
US20140199234A1 (en) 2013-01-16 2014-07-17 The Regents Of The University Of Michigan Bcl-2/bcl-xl inhibitors and therapeutic methods using the same
US20140275082A1 (en) 2013-03-14 2014-09-18 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2014165044A1 (fr) 2013-03-13 2014-10-09 Abbvie Inc. Procédés de préparation d'un agent induisant l'apoptose
CN104131034A (zh) 2014-06-19 2014-11-05 中山大学 一种嵌合载体及其制备方法和应用
WO2014189393A2 (fr) 2013-05-24 2014-11-27 Hampton Mark Barry Composés bioactifs
WO2015003816A2 (fr) 2013-07-12 2015-01-15 Helmholtz-Zentrum für Infektionsforschung GmbH Cystobactamides
US9034875B2 (en) 2009-05-26 2015-05-19 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2015085238A1 (fr) 2013-12-05 2015-06-11 The Regents Of The University Of California, A California Corporation Inhibiteurs de lpxc
WO2015158299A1 (fr) 2014-04-17 2015-10-22 沈华浩 Application de composé de sulfonamides à une maladie inflammatoire chronique des voies aériennes
WO2016025652A1 (fr) 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinaisons d'un inhibiteur de erk et d'un modulateur de la voie bcl-2 et méthodes associées
US20160220573A1 (en) 2015-02-03 2016-08-04 Gilead Sciences, Inc. Combination therapies for treating cancers
WO2016127135A1 (fr) 2015-02-06 2016-08-11 Unity Biotechnology, Inc. Composés et utilisations dans le traitement d' états associés à la sénescence
US20160243247A1 (en) 2014-12-23 2016-08-25 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
WO2016131100A1 (fr) 2015-02-18 2016-08-25 The Walter And Eliza Hall Institute Of Medical Research Méthodes de traitement de maladies infectieuses
WO2016172194A1 (fr) 2015-04-20 2016-10-27 Mayo Foundation For Medical Education And Research Procédés et matériaux pour détruire des cellules infectées par le vih
WO2016188935A1 (fr) 2015-05-26 2016-12-01 F. Hoffmann-La Roche Ag Traitement combiné d'un anticorps anti-cd20 avec un inhibiteur de bcl-2 et un inhibiteur de mdm2
WO2017017469A1 (fr) 2015-07-30 2017-02-02 The University Of Manchester Dérivés de diarylboron cycliques utilisés en tant qu'inhibiteurs d'inflammasome nlrp3
WO2017044720A1 (fr) 2015-09-11 2017-03-16 Navitor Pharmaceuticals, Inc. Analogues de la rapamycine et leurs utilisations
US20170087162A1 (en) 2013-05-22 2017-03-30 Children's Hospital Medical Center Combination therapy for mds
CN106565706A (zh) 2016-10-27 2017-04-19 广东东阳光药业有限公司 一种磺酰胺衍生物及其在药学中的应用
CN106608895A (zh) 2015-10-21 2017-05-03 苏州国匡医药科技有限公司 一类具有糖环结构的bcl-2选择性抑制剂及其用途
CN106749233A (zh) 2016-11-24 2017-05-31 中山大学 一类磺酰胺衍生物及其应用
WO2017101851A1 (fr) 2015-12-18 2017-06-22 Unity Biotechnology, Inc. Dérivés acylsulfonamide pour le traitement de maladies et de troubles associés à la sénescence
CN106957315A (zh) 2016-01-08 2017-07-18 中国人民解放军第二军医大学 N-取代苯磺酰基-氮杂吲哚氧基苯甲酰胺类化合物及其制备药物的用途
WO2017123616A1 (fr) 2016-01-11 2017-07-20 Merrimack Pharmaceuticals, Inc. Inhibition du lymphome 2 à cellules b (bcl -2) et des protéines apparentées
WO2017132474A1 (fr) 2016-01-30 2017-08-03 Newave Pharmaceutical Inc. Inhibiteurs de bcl-2
CN107089981A (zh) 2017-04-24 2017-08-25 杭州科耀医药科技有限公司 一种BCL‑2抑制剂Venetoclax的合成方法
WO2017184995A1 (fr) 2016-04-21 2017-10-26 Bioventures, Llc Composés induisant la dégradation de protéines anti-apoptotiques de la famille bcl-2 et utilisation de ces derniers
WO2017214491A1 (fr) 2016-06-09 2017-12-14 Blinkbio, Inc. Charges utiles thérapeutiques à base de silanol
WO2018026646A1 (fr) 2016-08-02 2018-02-08 Janssen Biotech, Inc. Traitement d'association pour cancers hématologiques
WO2018127130A1 (fr) 2017-01-07 2018-07-12 Shanghai Fochon Pharmaceutical Co., Ltd. Composés en tant qu'agents induisant l'apoptose sélective de bcl-2
WO2018146506A1 (fr) 2017-02-10 2018-08-16 Universitat Politècnica De València Dérivés thérapeutiques
WO2019210828A1 (fr) 2018-04-29 2019-11-07 Beigene, Ltd. Inhibiteurs de bcl-2
WO2020061216A1 (fr) 2018-09-18 2020-03-26 Signalchem Lifesciences Corporation Polythérapie pour traiter le cancer du sang
US20200339605A1 (en) 2018-01-10 2020-10-29 Recurium Ip Holdings, Llc Benzamide compounds

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2423029T3 (es) 2006-11-15 2013-09-17 Genentech, Inc. Compuestos de arilsulfonamida
US8563735B2 (en) 2008-12-05 2013-10-22 Abbvie Inc. Bcl-2-selective apoptosis-inducing agents for the treatment of cancer and immune diseases
TWI535712B (zh) 2010-08-06 2016-06-01 阿斯特捷利康公司 化合物
WO2016044331A1 (fr) * 2014-09-17 2016-03-24 Kalyra Pharmaceuticals, Inc. Composés bicycliques
UY36391A (es) * 2014-11-05 2016-06-01 Flexus Biosciences Inc Compuestos moduladores de la enzima indolamina 2,3-dioxigenasa (ido1), sus métodos de síntesis y composiciones farmacèuticas que las contienen

Patent Citations (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003096461A (ja) 2001-09-25 2003-04-03 Fuji Photo Film Co Ltd 液晶組成物およびそれを用いた液晶素子
WO2003089441A1 (fr) 2002-04-22 2003-10-30 Ibfb Gmbh Mercaptans tricycliques, leur procede de production et leur utilisation
US20060258657A1 (en) 2003-11-13 2006-11-16 Milan Bruncko Apoptosis promoters
WO2005049593A2 (fr) 2003-11-13 2005-06-02 Abbott Laboratories Promoteurs de l'apoptose contenant n-acylsulfonamide
WO2005049594A1 (fr) 2003-11-13 2005-06-02 Abbott Laboratories Promoteurs de l'apoptose contenant n-acylsulfonamide
US20060128706A1 (en) 2003-11-13 2006-06-15 Milan Bruncko Apoptosis promoters
US20070015787A1 (en) 2003-11-13 2007-01-18 Milan Bruncko Apoptosis promoters
WO2005079803A1 (fr) 2004-02-13 2005-09-01 Pfizer Products, Inc. Composes pour traitement des maladies cardio-vasculaires
WO2006093346A1 (fr) 2005-03-03 2006-09-08 Fujifilm Corporation Film d’acylate de cellulose, plaque polarisante et dispositif d’affichage a cristaux liquides
US20070027135A1 (en) 2005-05-12 2007-02-01 Milan Bruncko Apoptosis promoters
US20100227838A1 (en) 2005-05-12 2010-09-09 Abbott Laboratories Combination Therapy for Treating Cancer and Diagnostic Assays for Use Therein
WO2008008374A2 (fr) 2006-07-14 2008-01-17 Chemocentryx, Inc. Inhibiteurs de ccr2 et leurs procédés d'utilisation
WO2008030836A2 (fr) 2006-09-05 2008-03-13 Abbott Laboratories Traitement des maladies myéloprolifératives
WO2008033747A2 (fr) 2006-09-11 2008-03-20 Curis, Inc. Petites molécules multifonctionnelles servant d'agents anti-prolifératifs
WO2008064116A2 (fr) 2006-11-16 2008-05-29 Abbott Laboratories Procédé pour éviter ou traiter les rejets lors de transplantations d'organes, de cellules souches hématopoïétiques ou de moelle osseuse
US20080199873A1 (en) 2006-12-04 2008-08-21 Anderson Mark G Companion diagnostic assays for cancer therapy
WO2009036051A1 (fr) 2007-09-10 2009-03-19 Curis, Inc. Inhibiteurs de la bcl-2 contenant une fraction de liaison au zinc
WO2010065824A2 (fr) 2008-12-04 2010-06-10 Abbott Laboratories Agents induisant l'apoptose pour le traitement du cancer et de maladies immunes et auto-immunes
US9029404B2 (en) 2008-12-04 2015-05-12 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
US8952157B2 (en) 2008-12-04 2015-02-10 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
US20100160322A1 (en) 2008-12-04 2010-06-24 Abbott Laboratories Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
US8557983B2 (en) 2008-12-04 2013-10-15 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2010065865A2 (fr) 2008-12-05 2010-06-10 Abbott Laboratories Agents induisant l'apoptose sélectifs pour bcl-2 pour le traitement du cancer et des maladies immunes
WO2010083442A1 (fr) 2009-01-19 2010-07-22 Abbott Laboratories Agents induisant une apoptose pour le traitement du cancer et de maladies immunes et auto-immunes
US20100305122A1 (en) 2009-05-26 2010-12-02 Abbott Laboratories Apoptosis inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2010138588A2 (fr) 2009-05-26 2010-12-02 Abbott Laboratories Agents induisant l'apoptose, dans le traitement du cancer et de maladies immunes et auto-immunes
US20170158666A1 (en) 2009-05-26 2017-06-08 Abbvie Inc. Apoptosis inducing agents for the treatment of cancer and immune and autoimmune diseases
US9034875B2 (en) 2009-05-26 2015-05-19 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
US20100311751A1 (en) 2009-06-08 2010-12-09 Abbott Laboratories Solid dispersions containing an apoptosis-promoting agent
US20100323020A1 (en) 2009-06-18 2010-12-23 Abbott Laboratories Stable nanoparticulate drug suspension
WO2010148422A1 (fr) 2009-06-25 2010-12-29 Walter And Eliza Hall Institute Of Medical Research Composés et procédés de traitement d'infestations parasitaires
WO2011021038A1 (fr) 2009-08-20 2011-02-24 Karus Therapeutics Limited Composés hétérocycliques tricycliques en tant qu'inhibiteurs de phosphoinositide 3-kinase
CN101798292A (zh) 2010-03-29 2010-08-11 无锡好芳德药业有限公司 ABT-263衍生的新型Bcl-2蛋白抑制剂的制备
WO2011150016A1 (fr) 2010-05-26 2011-12-01 Abbott Laboratories Agents inducteurs d'apoptose pour le traitement du cancer et de maladies immunitaires et auto-immunes
US20120108590A1 (en) 2010-10-29 2012-05-03 Abbott Laboratories Melt-extruded solid dispersions containing an apoptosis-inducing agent
WO2012058392A1 (fr) 2010-10-29 2012-05-03 Abbott Laboratories Dispersions solides contenant un agent induisant l'apoptose
WO2012063085A2 (fr) 2010-11-11 2012-05-18 Redx Pharma Limited Dérivés de médicaments
US20120129853A1 (en) 2010-11-23 2012-05-24 Abbott Laboratories Methods of treatment using selective bcl-2 inhibitors
US20120189539A1 (en) 2011-01-25 2012-07-26 The Regents Of University Of Michigan Bcl-2/bcl-xl inhibitors and therapeutic methods using the same
WO2013185202A1 (fr) 2012-06-14 2013-12-19 Beta Pharma Canada Inc Inducteurs d'apoptose
US20140199234A1 (en) 2013-01-16 2014-07-17 The Regents Of The University Of Michigan Bcl-2/bcl-xl inhibitors and therapeutic methods using the same
WO2014113413A1 (fr) 2013-01-16 2014-07-24 The Regents Of The University Of Michigan Inhibiteurs de bcl-2bcl-xl et méthodes thérapeutiques les faisant intervenir
US20150336994A1 (en) 2013-01-16 2015-11-26 The Regents Of The University Of Michigan Bcl-2/bcl-xl inhibitors and therapeutic methods using the same
WO2014165044A1 (fr) 2013-03-13 2014-10-09 Abbvie Inc. Procédés de préparation d'un agent induisant l'apoptose
US20140275082A1 (en) 2013-03-14 2014-09-18 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
US20170087162A1 (en) 2013-05-22 2017-03-30 Children's Hospital Medical Center Combination therapy for mds
WO2014189393A2 (fr) 2013-05-24 2014-11-27 Hampton Mark Barry Composés bioactifs
WO2015003816A2 (fr) 2013-07-12 2015-01-15 Helmholtz-Zentrum für Infektionsforschung GmbH Cystobactamides
WO2015085238A1 (fr) 2013-12-05 2015-06-11 The Regents Of The University Of California, A California Corporation Inhibiteurs de lpxc
WO2015158299A1 (fr) 2014-04-17 2015-10-22 沈华浩 Application de composé de sulfonamides à une maladie inflammatoire chronique des voies aériennes
CN104131034A (zh) 2014-06-19 2014-11-05 中山大学 一种嵌合载体及其制备方法和应用
WO2016025652A1 (fr) 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinaisons d'un inhibiteur de erk et d'un modulateur de la voie bcl-2 et méthodes associées
US20160243247A1 (en) 2014-12-23 2016-08-25 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
US20160220573A1 (en) 2015-02-03 2016-08-04 Gilead Sciences, Inc. Combination therapies for treating cancers
WO2016127135A1 (fr) 2015-02-06 2016-08-11 Unity Biotechnology, Inc. Composés et utilisations dans le traitement d' états associés à la sénescence
WO2016131100A1 (fr) 2015-02-18 2016-08-25 The Walter And Eliza Hall Institute Of Medical Research Méthodes de traitement de maladies infectieuses
WO2016172194A1 (fr) 2015-04-20 2016-10-27 Mayo Foundation For Medical Education And Research Procédés et matériaux pour détruire des cellules infectées par le vih
WO2016188935A1 (fr) 2015-05-26 2016-12-01 F. Hoffmann-La Roche Ag Traitement combiné d'un anticorps anti-cd20 avec un inhibiteur de bcl-2 et un inhibiteur de mdm2
WO2017017469A1 (fr) 2015-07-30 2017-02-02 The University Of Manchester Dérivés de diarylboron cycliques utilisés en tant qu'inhibiteurs d'inflammasome nlrp3
WO2017044720A1 (fr) 2015-09-11 2017-03-16 Navitor Pharmaceuticals, Inc. Analogues de la rapamycine et leurs utilisations
CN106608895A (zh) 2015-10-21 2017-05-03 苏州国匡医药科技有限公司 一类具有糖环结构的bcl-2选择性抑制剂及其用途
WO2017101851A1 (fr) 2015-12-18 2017-06-22 Unity Biotechnology, Inc. Dérivés acylsulfonamide pour le traitement de maladies et de troubles associés à la sénescence
CN106957315A (zh) 2016-01-08 2017-07-18 中国人民解放军第二军医大学 N-取代苯磺酰基-氮杂吲哚氧基苯甲酰胺类化合物及其制备药物的用途
WO2017123616A1 (fr) 2016-01-11 2017-07-20 Merrimack Pharmaceuticals, Inc. Inhibition du lymphome 2 à cellules b (bcl -2) et des protéines apparentées
WO2017132474A1 (fr) 2016-01-30 2017-08-03 Newave Pharmaceutical Inc. Inhibiteurs de bcl-2
WO2017184995A1 (fr) 2016-04-21 2017-10-26 Bioventures, Llc Composés induisant la dégradation de protéines anti-apoptotiques de la famille bcl-2 et utilisation de ces derniers
WO2017214491A1 (fr) 2016-06-09 2017-12-14 Blinkbio, Inc. Charges utiles thérapeutiques à base de silanol
WO2018026646A1 (fr) 2016-08-02 2018-02-08 Janssen Biotech, Inc. Traitement d'association pour cancers hématologiques
CN106565706A (zh) 2016-10-27 2017-04-19 广东东阳光药业有限公司 一种磺酰胺衍生物及其在药学中的应用
CN106749233A (zh) 2016-11-24 2017-05-31 中山大学 一类磺酰胺衍生物及其应用
WO2018127130A1 (fr) 2017-01-07 2018-07-12 Shanghai Fochon Pharmaceutical Co., Ltd. Composés en tant qu'agents induisant l'apoptose sélective de bcl-2
US11053239B2 (en) 2017-01-07 2021-07-06 Fochon Pharmaceuticals, Ltd. Compounds as BLC-2-selective apoptosis-inducing agents
WO2018146506A1 (fr) 2017-02-10 2018-08-16 Universitat Politècnica De València Dérivés thérapeutiques
CN107089981A (zh) 2017-04-24 2017-08-25 杭州科耀医药科技有限公司 一种BCL‑2抑制剂Venetoclax的合成方法
US20200339605A1 (en) 2018-01-10 2020-10-29 Recurium Ip Holdings, Llc Benzamide compounds
US20200338071A1 (en) 2018-01-10 2020-10-29 Recurium Ip Holdings, Llc Benzamide compounds
WO2019210828A1 (fr) 2018-04-29 2019-11-07 Beigene, Ltd. Inhibiteurs de bcl-2
WO2020061216A1 (fr) 2018-09-18 2020-03-26 Signalchem Lifesciences Corporation Polythérapie pour traiter le cancer du sang

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
Aguilar, A. et al, "A Potent and Highly Efficacious Bcl-2/Bcl-xL Inhibitor" Journal of Medicinal Chemistry (2013), 56(7), 3048-3067.
Almerico, A. et al, "3D-QSAR pharmacophore modeling and in silico screening of new Bcl-xl inhibitors" European Journal of Medicinal Chemistry, (2010),(11), 4774-4782.
Anderson, Y. et al, "Improving Nonspecific Binding and Solubility: Bicyloalkyl Groups and Cubanese as para-Phenyl Biososteres" ChemMedChem (2017), 12, 590-598.
Bai, L. et al, "BM-1197: a novel and specific Bcl-2/Bcl-xL inhibitor inducing complete and long lasting tumor regression in vivo" PLoS One (2014), 9(6), e99404/1-e99404/13, 13 pp.
Bernardo P. et al, "Structure-Activity Reiationship Studies of Phenanthridine-Based Bcl-XL Inhibitors" J Med Chem, (2008), 51(21), 6699-6710.
Bruncko, M. et al., "Studies Leading to Potent, Dual Inhibitors of Bcl-2 and Bcl-Xl" Journal of Medicinal Chemistry 2007 50 (4), 641-662.
Chen, J. et al, "Structure-Based Digcovery of BM-957 as a Potent Small-Molecule Inhibitor of Bcl-2 and Bcl-xL Capable of Achieving Complete Tumor Regression" Journal of Medicinal Chemistry (2012), 55(19), 8502-8514.
Extended European search report for EP 19738922.4, dated Aug. 12, 2021.
Giedt R. et al, "Imaging Cellular Distribution of Bcl Inhibitors Using Small Molecule Drug Conjugates" Bioconjugate Chemistry (2014), 25(11), 2081-2085
Hou, X. et al, "3D-QSAR Study on a series of Bcl-2 protein inhibitors using comparative molecular field analysis" Protein & Peptide Letters (2011), 18(5), 440-449.
International Search Report for PCT/US2019/012704, filed Jan. 8, 2019.
Kastritis, P. et al, "A Biophysical Model for Predicting the Binding Affinity of Protein-Protein Interactions Inhibitors" Journal of Chemical Information and Modeling (2014), 54(3), 826-836.
Lee, E. et al, "Conformational Changes in Bcl-2 Pro-survival Proteins Determine their Capacity to Bing Ligands" Journal of Biological Chemistry (2009), 284(44), 30508-30517.
Mabrouk, M. "Discovering best candidates for hepatocellular carcinoma(HCC) by in-silico techniques and tools" Int. J. Bioinfo. Res. and Appl.,(2012), 8(1/2), 141-152.
Magee, Thomas V., "Progress in discovery of small-molecule modulators of protein-protein interactions via fragment screening" Bioorganic & Medicinal Chemistry Letters, 2015, 25(12), 2461.
Park, C-M. at al, "Discovery of an Orally Bioavailable Small Molecule Inhibitor of Prosurvival B-Cell Lymphoma 2 Proteins" J Med Chem, (2008), 51(21), 6902-6915.
Shipra, G. et al., "Identification of Novel Potent Inhibitors Against Bcl-xL Anti-apoptotic Protein Using Docking Studies" vol. 19, No. 12, 2012, pp. 1302-1317.
Varnes, J. et al, "Towards the next generation of dual Bcl-2/Bcl-xL inhibitors" Bioorganic & Medicinal Chemistry Letters (2014), 24(14), 3026-3033.
Written Opinion and Search Report for Singapore Application No. 11202005785X, dated Jan. 6, 2022.
Ye, L. et al, "The small-molecule compound BM-1197 inhibits the antiapoptotic regulators Bcl-2/Bcl-xL and triggers apoptotic cell death in human colorectal cancer cells" Tumor Biology (2015), 36(5), 3447-3455.
Zhou, H. et al, "Design of Bcl-2 and Bcl-xL Inhibitors with Subnanomolar Binding Affinities Based upon a New Scaffold [Erratum to document cited in CA157:045136]" Journal of Medicinal Chemistry (2012), 55(12), 5987.
Zhou, H. et al, "Structure-Based Design of Potent Bcl-2/Bcl-xL Inhibitors with Strong in Vivo Antitumor Activity" Journal of Medicinial Chemistry (2012), 55(13), 6149-6161.

Also Published As

Publication number Publication date
IL275763A (en) 2020-08-31
JP2021510376A (ja) 2021-04-22
JP2024001052A (ja) 2024-01-09
AR114998A1 (es) 2020-11-18
CN111801327B (zh) 2024-02-09
AR114950A1 (es) 2020-11-11
US20220226314A1 (en) 2022-07-21
WO2019139899A8 (fr) 2020-07-16
CA3087263A1 (fr) 2019-07-18
RU2020120928A (ru) 2022-02-25
SG11202005785XA (en) 2020-07-29
EP3737672A4 (fr) 2021-09-15
BR112020014169A2 (pt) 2020-12-08
EP3737672A1 (fr) 2020-11-18
JP7355741B2 (ja) 2023-10-03
CA3087261A1 (fr) 2019-07-18
RU2020120929A (ru) 2022-02-25
MX2023009185A (es) 2023-08-21
WO2019139899A1 (fr) 2019-07-18
JP2021510163A (ja) 2021-04-15
MX2020007392A (es) 2020-10-14
TW201934538A (zh) 2019-09-01
US11813260B1 (en) 2023-11-14
EP3740487A1 (fr) 2020-11-25
MX2020007388A (es) 2020-10-12
BR112020014151A2 (pt) 2020-12-08
US11590126B2 (en) 2023-02-28
WO2019139902A1 (fr) 2019-07-18
TW201934550A (zh) 2019-09-01
US20210009543A1 (en) 2021-01-14
RU2020120933A (ru) 2022-02-10
TWI829664B (zh) 2024-01-21
IL275761A (en) 2020-08-31
AR114949A1 (es) 2020-11-11
CN111801327A (zh) 2020-10-20
KR20200108301A (ko) 2020-09-17
SG11202005985PA (en) 2020-07-29
CA3087262A1 (fr) 2019-07-18
AU2019207616A1 (en) 2020-07-09
AU2019207608B2 (en) 2024-03-28
WO2019139907A1 (fr) 2019-07-18
AU2019207608A1 (en) 2020-07-09
NZ765825A (en) 2023-12-22
KR20200108302A (ko) 2020-09-17
AU2019207611A1 (en) 2020-07-09
US20200338071A1 (en) 2020-10-29
JP2021510165A (ja) 2021-04-15
AR114948A1 (es) 2020-11-11
US20240180899A1 (en) 2024-06-06
CN111801320A (zh) 2020-10-20
EP3740487A4 (fr) 2021-11-10
TW201930317A (zh) 2019-08-01
IL275762A (en) 2020-08-31
EP3737681A4 (fr) 2022-01-12
CN111788203A (zh) 2020-10-16
EP3737681A1 (fr) 2020-11-18
WO2019139900A1 (fr) 2019-07-18
US11813259B2 (en) 2023-11-14
US20200339605A1 (en) 2020-10-29
SG11202005784YA (en) 2020-07-29
CN118084905A (zh) 2024-05-28
US11318134B2 (en) 2022-05-03
MX2020007014A (es) 2020-11-11
KR20200108298A (ko) 2020-09-17
BR112020014160A2 (pt) 2020-12-08
TW201930302A (zh) 2019-08-01
CN118084904A (zh) 2024-05-28

Similar Documents

Publication Publication Date Title
US11813260B1 (en) Benzamide compounds
US20220273666A1 (en) Nanoparticle formulation of bcl-2 inhibitor
RU2801647C2 (ru) Бензамидные соединения
AU2024203833A1 (en) Benzamide compounds
TW202417442A (zh) 苯甲醯胺化合物

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

AS Assignment

Owner name: RECURIUM IP HOLDINGS, LLC, CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:ZENO ROYALTIES & MILESTONES, LLC;REEL/FRAME:053144/0425

Effective date: 20190904

Owner name: ZENO ROYALTIES & MILESTONES, LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ZENO MANAGEMENT, INC.;REEL/FRAME:053142/0819

Effective date: 20190710

Owner name: ZENO MANAGEMENT, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PINCHMAN, JOSEPH ROBERT;HUANG, PETER QINHUA;BUNKER, KEVIN DUANE;AND OTHERS;REEL/FRAME:053142/0801

Effective date: 20190807

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: AWAITING TC RESP., ISSUE FEE NOT PAID

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction